US20070213291A1 - Therapeutically useful synthetic oligonucleotides - Google Patents

Therapeutically useful synthetic oligonucleotides Download PDF

Info

Publication number
US20070213291A1
US20070213291A1 US11/641,542 US64154206A US2007213291A1 US 20070213291 A1 US20070213291 A1 US 20070213291A1 US 64154206 A US64154206 A US 64154206A US 2007213291 A1 US2007213291 A1 US 2007213291A1
Authority
US
United States
Prior art keywords
seq
bases
cells
group
cancer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/641,542
Inventor
Nigel Phillips
Maria Filion
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US11/641,542 priority Critical patent/US20070213291A1/en
Publication of US20070213291A1 publication Critical patent/US20070213291A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7135Compounds containing heavy metals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/117Nucleic acids having immunomodulatory properties, e.g. containing CpG-motifs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55561CpG containing adjuvants; Oligonucleotide containing adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/18Type of nucleic acid acting by a non-sequence specific mechanism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3513Protein; Peptide

Definitions

  • the present invention relates to an oligonucleotide composition for the treatment of cancer.
  • Cancer is an aberrant net accumulation of atypical cells, which can result from an excess of proliferation, an insufficiency of cell death, or a combination of the two.
  • Proliferation is the culmination of a cell's progression through the cell cycle resulting in the division of one cell into two cells.
  • the 5 major phases of the cell cycle are G 0 , G 1 , S, G 2 and M.
  • G 0 phase cells are quiescent. Most cells in the body, at any one time, are in this stage.
  • G 1 phase cells, responding to signals to divide, produce the RNA and the proteins necessary for DNA synthesis.
  • SE early S-phase
  • SM middle S-phase
  • SL late S-phase
  • the cells replicate their DNA.
  • proteins are elaborated in preparation for cell division.
  • the mitotic (M) phase the cell divides into two daughter cells. Alterations in cell cycle progression occur in all cancers and may result from over-expression of genes, mutation of regulatory genes, or abrogation of DNA damage checkpoints (Hochhauser D. Anti-Cancer Chemotherapeutic Agents 8:903, 1997).
  • cellular senescence is a programmed cell death response leading to growth arrest of cells (Dimri et al. Proc. Natl. Acad. Sci. USA 92:20, 1995). DNA damage, exposure of colon, breast and ovarian cancer cells to toposiomerase inhibitors and exposure of nasopharyngeal cancer cells to cisplatin are reported to prevent proliferation of these cells by induction of senescence (Wang et al. Cancer Res. 58:5019, 1998; Poele et al. Br. J. Cancer 80:9, 1999).
  • Synthetic oligonucleotides are polyanionic sequences that are internalized in cells (Vlassov et al. Biochim. Biophys. Acta 1197:95, 1994). Synthetic oligonucleotides are reported that bind selectively to nucleic acids (Wagner, R. Nature: 372:333, 1994), to specific cellular proteins (Bates et al. J. Biol. Chem. 274:26369, 1999) and to specific nuclear proteins Scaggiante et al. Eur. J. Biochem. 252:207, 1998) to inhibit proliferation of cancer cells.
  • Synthetic 27 base sequences containing guanine (G) and variable amounts of thymine (T) (oligonucleotide GTn), wherein n is ⁇ 1 or ⁇ 7 and wherein the number of bases is ⁇ 20 are reported to inhibit growth of cancer cell lines by sequence specific binding to a 45 kDa nuclear protein, whereas GTn, wherein the number of bases is ⁇ 20, are reported to be inactive against cancer cell lines (Morassutti et al. Nucleosides and Nucleotides 18:1711, 1999).
  • Apoptosis is an active cellular death process characterized by distinctive morphological changes that include condensation of nuclear chromatin, cell shrinkage, nuclear disintegration, plasma membrane blebbing, and the formation of membrane-bound apoptotic bodies (Wyllie et al. Int. Rev. Cytol. 68:251, 1980).
  • a molecular hallmark of apoptosis is degradation of the cell's nuclear DNA into oligonucleosomal-length fragments as the result of activation of endogenous endonucleases (Wyllie A. Nature 284:555, 1980).
  • Caspases (cysteine-aspartyl-specific proteases) have been implicated as key enzymes in the execution of the late stage of apoptosis.
  • the caspase family consists of at least fourteen related cysteine aspartyl proteases. All the caspases contain a conserved QACXG (where X is R, Q or G) pentapeptide activesite motif (Cohen G. Biochim. Biophys. Acta 1366:139, 1997).
  • a number of caspases are synthesized as inactive proenzymes that are activated following cleavage at caspase specific cleavage sites (Cohen G. Biochim. Biophys. Acta 1366:139, 1997) or as inactive enzymes that require association with regulatory molecules for activation (Stennicke et al. J. Biol. Chem. 274:8359, 1999).
  • caspases are involved in activation and proliferation of B and T lymphocytes, in cytokine maturation during inflammation, in differentiation of progenitor cells during erythropoiesis and in development of lens fiber (Fadeel et al. Leukemia 14:1514, 2000).
  • caspase 3 is processed during activation of B lymphocytes and of CD4 (+), CD8 (+), CD45RA(+) and CD45RO (+) subsets of T lymphocytes (Alam et al. J. Exp. Med. 190:1879, 1999).
  • T lymphocytes stimulation of T lymphocytes by mitogens and by interleukin-2 is associated with activation of the caspase pathway and with cleavage of PARP (Wilheim et al. Eur. J. Immunol. 28:891, 1998).
  • caspase 3 activity is necessary for the release of IL-2 by activated T lymphocytes (Posmantur et al. Exp. Cell Res. 244:302, 1998) and for the processing and maturation of the pro-inflammatory cytokine interleukin-16 (Zhang et al. J. Biol. Chem. 273:1144, 1998).
  • GATA-1 a nuclear regulatory protein crucial for the maturation of erythroid precursors
  • Cytolysis is the complete or partial destruction of a cell and is mediated by the immune system.
  • Activated macrophages and monocytes produce bioactive molecules that include, but are not limited to cytokines.
  • Cytokines include, but are not limited to, interleukin (IL)-1, IL-1 beta, IL-6, IL-10, IL-12, and TNF-alpha.
  • IL-1beta reduces bone marrow cell sensitivity to cytoreductive drugs, to radiation and to in vitro tumor cell purging with drugs in autologous bone marrow transplantation (Dalmau et al. Bone Marrow Transplant. 12:551, 1993).
  • IL-6 induces B cell differentiation, stimulates IgG secretion (Taga et al. J. Exp. Med. 166:967, 1987), induces cytotoxic T cell differentiation (Lee et al. Vaccine 17:490, 1999), promotes megakaryocyte maturation (Ishibashi et al. Proc. Natl. Acad. Sci. USA 86:8953, 1989) and functions both as an anti proliferative factor (Mori et al. Biochem. Biophys. Res. Comm. 257:609, 1999; Alexandroff et al. Biochem. Soc. Trans. 25:270, 1997; Takizawa et al. Cancer Res. 53:18, 1993: Novick et al.
  • Cytokine 4:6, 1992 and as a pro-proliferative factor (Okamoto et al. Cancer Res. 57:141, 1997; Okamoto et al. Int. J. Cancer 72:149, 1997; Chiu et al. Clin. Cancer Res. 2:215, 1996; Lu et al. Clin. Cancer Res. 2:1417, 1996) for cancer cells.
  • IL-10 enhances the effectiveness of vaccines in murine tumor models (Kauffman et al. J. Immunother. 22: 489, 1999) and up-regulates anti-cancer autoreactive T cell responses (Alleva et al. Immunobiol. 192:155, 1995).
  • IL-12 alone and in combination with other cytokines, promotes the maturation of leukocytes and induces the secretion of interferon-gamma.
  • IL-12 is reported to have anti-cancer activity (Stine et al. Annals NY Academy of Science 795:420, 1996; Chen et al. Journal of Immunol. 159:351, 1997) including, but not limited to, activation of specific cytolytic T-lymphocytes, activation of natural killer (NK) cells and induction of the anti-angiogenic proteins IP-10 and MiG.
  • TNF-alpha causes necrosis of solid tumors (Porter et al. Trends in Biotech. 9:158, 1991), sensitizes cancer cells to gamma irradiation-induced apoptosis (Kimura et al. Cancer Res. 59:1606, 1999) and protects bone marrow precursor cells from the effects of antineoplastic agents (Dalmau et al. Bone Marrow Transplant. 12:551, 1993).
  • compositions and methods that induce cell cycle arrest in cancer cells, inhibit proliferation of cancer cells, activate caspases in cancer cells, induce apoptosis in cancer cells and stimulate cytokine production by immune system cells.
  • the present invention fulfills this need by providing a composition and method comprising a 2 to 20 base 3′-OH, 5′-OH synthetic oligonucleotide (hereinafter “sequence”) selected from the group consisting of (G x T y ) n , (T y G x ) n , a(G x T y ) n , a(T y G x ) n , (G x T y ) n b, (T y G x ) n b, a(G x T y ) n b, a(T y G x ) n b, wherein x and y is an integer between 1 and 7, n is an integer between 1 and 12, a and b are one or more As, Cs, Gs or Ts, and wherein the sequence induces a response selected from the group consisting of induction of cell cycle arrest, inhibition of proliferation, activation of caspases and induction
  • a composition comprising a sequence and a pharmaceutically acceptable carrier is administered to an animal, including a human, having cancer in an amount effective to treat the cancer in the animal.
  • the unexpected and surprising ability of the sequence to induce cell cycle arrest, inhibit proliferation, activate caspases and induce apoptosis and in cancer cells and to stimulate cyotkine production by immune system cells addresses a long felt unfulfilled need in the medical arts and provides an important benefit for animals, including humans.
  • an object of the present invention is to provide a composition and method effective to treat a disease in an animal, including a human.
  • Another object of the present invention is to provide a composition and method effective to treat a cancer.
  • Another object of the present invention is to provide a composition and method that induces senescence in cells.
  • Another object of the present invention is to provide a composition and method that induces cell cycle arrest in cells.
  • Another object of the present invention is to provide a composition and method that induces cell cycle arrest in cancer cells.
  • Another object of the present invention is to provide a composition and method that inhibits proliferation of cells.
  • Another object of the present invention is to provide a composition and method that inhibits proliferation of cancer cells.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cells.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of Fas.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of TNFR1.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of p53/p21.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of p21/waf-1/CIP.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of p15 ink4B .
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of p16 ink4 .
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of caspase 3.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of TGF-beta 1 receptor.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of hormone dependence.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of drug resistance.
  • Another object of the present invention is to provide a composition and method that activates caspases in cells.
  • Another object of the present invention is to provide a composition and method that activates caspases in cancer cells.
  • Another object of the present invention is to provide a composition and method to treat an autoimmune disease.
  • Another object of the present invention is to provide a composition and method to treat a lymphoproliferative disease.
  • Another object of the present invention is to provide a composition and method to treat an infection.
  • Another object of the present invention is to provide a composition and method to treat an inflammation.
  • Another object of the present invention is to provide a composition and method to modulate T- or B-cell activation.
  • Another object of the present invention is to provide a composition and method to modulate progenitor cell maturation.
  • Another object of the present invention is to provide a composition and method to modulate erythropoiesis.
  • Another object of the present invention is to provide a composition and method to modulate transcription factors in cells.
  • Another object of the present invention is to provide a composition and method that potentiates the effect of other therapeutic agents on cells.
  • Another object of the present invention is to provide a composition and method that potentiates the effect of chemotherapeutic agents on cancer cells.
  • Another object of the present invention is to provide a composition and method that potentiates the anti-neoplastic effect of radiation.
  • Another object of the present invention is to provide a composition and method that stimulates cytokine production by cells of the immune system.
  • Another object of the present invention is to provide a composition and method that stimulates IL-1 beta production by cells of the immune system.
  • Another object of the present invention is to provide a composition and method that stimulates IL-6 production by cells of the immune system.
  • Another object of the present invention is to provide a composition and method that stimulates IL-10 production by cells of the immune system.
  • Another object of the present invention is to provide a composition and method that stimulates IL-12 production by cells of the immune system.
  • Another object of the present invention is to provide a composition and method that stimulates TNF- ⁇ production by cells of the immune system.
  • Another object of the present invention is to provide a composition that is simple to prepare.
  • Another object of the present invention is to provide a composition that is minimally toxic to the recipient.
  • FIG. 1 Fluorescence [Fluo-3-AM] of Jurkat human T leukemia cells incubated with 0 ⁇ g/ml and 100 ⁇ g/ml of 6 base GT SEQ ID NO:25.
  • FIG. 2 Caspase 3 activation in Jurkat human T cell leukemia cells incubated without 0 ⁇ g/ml and 100 ⁇ g/ml of GT SEQ ID NOs:66, 67, 81, 82 and 83 measured cytometrically (A) and colorimetrically (B).
  • FIG. 3 Caspase activation in Jurkat human T cell leukemia cells.
  • A Caspase 3 activation in cells incubated with 0 ⁇ g/ml and 100 ⁇ g/ml of 6 base GT SEQ ID NO:25;
  • B Caspase 7 activation(a) and PARP content (b) in cells incubated with 0 ⁇ g/ml and 100 ⁇ g/ml of 6 base GT SEQ ID NO:25.
  • the present invention provides a composition
  • a composition comprising a 2 to 20 base 3′-OH, 5′OH synthetic oligonucleotide (sequence) selected from the group consisting of (G x T y ) n , (T y G x ) n , a(G x T y ) n , a(T y G x ) n , (G x T y ) n b, (T y G x ) n b, a(G x T y ) n b, a(T y G x ) n b, wherein x and y is an integer between 1 and 7, n is an integer between 1 and 12, a and b are one or more As, Cs, Gs or Ts, wherein the sequence induces a response selected from the group consisting of induction of cell cycle arrest, inhibition of proliferation, activation of caspases and induction of apoptosis in
  • a composition comprising a sequence and a pharmaceutically acceptable carrier is administered to an animal, including a human, having cancer in an amount effective to treat the cancer in the animal, including the human.
  • the unexpected and surprising ability of the sequence to induce cell cycle arrest, inhibit proliferation, induce apoptosis and activate caspases in cancer cells and to stimulate cytokine production by immune system cells addresses a long felt unfulfilled need in the medical arts and provides an important benefit for animals, including humans.
  • sequence refers to a 2 to 20 base 3′-OH, 5′-OH synthetic oligonucleotide comprising A, C, G and T bases.
  • GT As used herein, the abbreviations “GT”, “AG” “CG”, “GG”, “AGT” and “CGT” refer to sequences comprising the named bases synthesized in any order.
  • response refers to induction of cell cycle arrest, inhibition of proliferation, activation of caspases and induction of apoptosis, in cancer cells and stimulation of cytokine production by immune system cells.
  • therapeutic treatment and “amount effective to” refer to an amount of a sequence effective to induce cell cycle arrest, inhibit proliferation, activate caspases and induce apoptosis in cancer cells and stimulate cytokine production by immune system cells.
  • solid tumor models refer to primary or secondary carcinomas or sarcomas.
  • chemotherapeutic is any agent approved by a regulatory agency of a country or a state government or listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia to treat cancer in an animal, including a human.
  • anti-plastic refers to preventing the development, maturation, proliferation or spread of cancer cells.
  • the word “potentiates” refers to a degree of synergism that is greater than the additive activity of each agent.
  • the word “synergism” refers to the coordinated action of two or more agents.
  • Administration of an effective amount of a sequence of the present invention to an animal, including a human, is a therapeutic treatment that prevents, treats or eliminates a disease including, but not limited to, cancer, rheumatoid arthritis, lympho-proliferative disorders and asthma.
  • Cancers include, but are not limited to, squamous cell carcinoma, fibrosarcoma, sarcoid carcinoma, melanoma, mammary cancer, lung cancer, colorectal cancer, renal cancer, osteosarcoma, cutaneous melanoma, basal cell carcinoma, pancreatic cancer, bladder cancer, brain cancer, ovarian cancer, prostate cancer, leukemia, lymphoma and metastases derived therefrom.
  • the therapeutic effectiveness of a sequence may be increased by methods including, but not limited to, chemically modifying the base, sugar or phosphate backbone, chemically supplementing or biotechnologically amplifying the sequences using bacterial plasmids containing the appropriate sequences, complexing the sequences to biological or chemical carriers or coupling the sequences to tissue-type or cell-type directed ligands or antibodies
  • compositions comprising one or more sequences and a pharmaceutically acceptable carrier are prepared by uniformly and intimately bringing into association the sequence and the pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers include liquid carriers, solid carriers or both.
  • Liquid carriers are aqueous carriers, non-aqueous carriers or both and include, but are not limited to, aqueous suspensions, oil emulsions, water in oil emulsions, water-in-oil-in-water emulsions, site-specific emulsions, long-residence emulsions, sticky-emulsions, microemulsions and nanoemulsions.
  • Solid carriers are biological carriers, chemical carriers or both and include, but are not limited to, viral vector systems, particles, microparticles, nanoparticles, microspheres, nanospheres, minipumps, bacterial cell wall extracts and biodegradable or non-biodegradable natural or synthetic polymers that allow for sustained release of the sequences.
  • Methods used to complex a sequence to a solid carrier include, but are not limited to, direct adsorption to the surface of the solid carrier; covalent coupling to the surface of the solid carrier, either directly or via a linking moiety; and covalent coupling to the polymer used to make the solid carrier.
  • a sequence can be stabilized by the addition of non-ionic or ionic polymers such as polyoxyethylenesorbitan monooleates (Tweens) or hyaluronic acid.
  • Preferred aqueous carriers include, but are not limited to, water, saline and pharmaceutically acceptable buffers.
  • Preferred non-aqueous carriers include, but are not limited to, a mineral oil and a neutral oil and mixtures thereof.
  • excipients may be included regardless of the pharmaceutically acceptable carrier used to present the sequence to the responding cells. These excipients include, but are not limited to, anti-oxidants, buffers, and bacteriostats, and may include suspending agents and thickening agents.
  • One or more sequences may be administered alone, or in combination with other therapeutic modalities including, but not limited to, chemotherapeutic agents, immunotherapeutic agents, antimicrobial agents, antiviral agents or in combination with radiation therapy.
  • Chemotherapeutic agents include, but are not limited to, anti-metabolites, DNA damaging, microtubule destabilizing, microtubule stabilizing, actin depolymerizing, growth inhibiting, topoisomerase inhibiting, HMG-CoA inhibiting, purine inhibiting, pyrimidine inhibiting, metaloproteinase inhibiting, CDK inhibiting, angiogenesis inhibiting and differentiation enhancing agents.
  • Routes of administration include, but are not limited to, oral, topical, subcutaneous, transdermal, subdermal, intramuscular, intra-peritoneal, intra-vesical, intra-articular, intra-arterial, intra-venous, intra-dermal, intra-cranial, intra-lesional, intra-tumoral, intra-ocular, intra-pulmonary, intra-spinal, intra-prostatic, placement within cavities of the body, nasal inhalation, pulmonary inhalation, impression into skin and electrocorporation.
  • the volume per dose is preferably about 0.001 to 100 ml per dose, more preferably about 0.01 to 50 ml per dose and most preferably about 0.1 to 30 ml per dose.
  • the amount of sequence administered per dose is from about 0.001 to 100 mg/kg, more preferably from about 0.01 to 10 mg/kg and most preferably from about 0.1 to 5 mg/kg.
  • the sequence or sequence plus a therapeutic agent can be administered in a single dose treatment, in multiple dose treatments or continuously infused on a schedule and over a period of time appropriate to the disease being treated, the condition of the recipient and the route of administration.
  • the sequence can be administered before, at the same time as, or after administration of the therapeutic agent.
  • a sequence in combination with chemotherapeutic agent is administered to an animal having cancer in an amount effective to potentiate the anti-neoplastic effect of the chemotherapeutic agent.
  • the amount of therapeutic agent administered per dose is from about 0.001 to 1000 mg/m 2 or from about 0.01 to 1000 mg/kg, more preferably from about 0.01 to 500 mg/m 2 or from about 0.01 to 500 mg/kg and most preferably from about 0.1 to 100 mg/m 2 or from about 0.1 to 100 mg/kg.
  • sequence and the particular therapeutic agent administered should be decided by the practitioner using methods known to those skilled in the art and will depend on the type of cancer, the severity of the cancer, the location of the cancer and other clinical factors such as the size, weight and physical condition of the recipient.
  • in vitro assays may optionally be employed to help identify optimal ranges for sequence administration and for sequence plus therapeutic agent administration.
  • sequences of the present invention form a new family of structures and that they do not function as antisense RNAs, antisense DNAs, triple helix forming DNAs, telomerase inhibitors, transcription activators or transcription inhibitors.
  • Phosphodiester and phosphorothioate sequences were prepared by HUKABEL Scientific Ltd, (Montréal, Québec, Canada) using the EXPEDITETM 8900 automated DNA synthesis system (PersSeptive Biosystems, Inc., Farminghan, Mass.) and by Sigma-Genosys (Woodlands, Tex.) using Abacus Segmented Synthesis Technology. Unless stated otherwise, the sequences used were phosphodiester sequences. Unless stated otherwise, immediately prior to use, the sequences were dispersed in autoclaved deionized water or in an autoclaved pharmaceutically acceptable buffer such as, but not limited to, saline.
  • Cells were seeded in 6 (1 ml/well), 24 (0.5 ml/well) or 96 (0.1 ml/well) well flat-bottom microplates and were maintained at 37° C. in a 5% CO 2 atmosphere. Unless stated otherwise, 2.5 ⁇ 10 5 cells/ml were incubated with 0 ⁇ g/ml (control) and 100 ⁇ g/ml (treated) of 2 to 45 base sequences containing A, C, G and T for 48 h.
  • MTT dimethylthiazol-diphenyl-tetrazolium
  • Jurkat human leukemia T cells are an atypical multi-drug resistant human suspension tumor cell model.
  • Jurkat cells were incubated with 27 base GT and CT sequences (Table 2).
  • TABLE 2 % inhibition of Jurkat human leukemia T cell proliferation SEQUENCE % INHIBITION GTGTGTGTGTGTGTGTGTGTGTG-(GT) 13 G 51 SEQ ID NO:1-(27 bases) GGGTGGGTGGGTGGGTGGGTGGG-(G 3 T) 6 G 3 23 SEQ ID NO:2-(27 bases) GGGGGTGGGGGTGGGTGGGTGGG-(G 5 T) 4 G 3 24 SEQ ID NO:3-(27 bases) GGGGGGGTGGGGGGGTGGGGGGGTGG-(G 7 T) 3 G 3 11 SEQ ID NO:4-(27 bases) TGTGTGTGTGTGTGTGTGTGTG-(TG) 13 T 45 SEQ ID NO:5-(27 bases) TCTCTCTCTCTCTCTCTCTCTCT-(TC)
  • Jurkat T cells were incubated with 6 base GT, AG, CG, GG, AGT and CGT sequences (Table 4). TABLE 4 % inhibition Jurkat human leukemia T cell proliferation SEQUENCE % INHIBITION TGTGTG-(TG) 3 36 SEQ ID NO:9-(6 bases) GTGTGT-(GT) 3 48 SEQ ID NO:10-(6 bases) TTTGTT-TT(TG) 1 TT 31 SEQ ID NO:23-(6 bases) GGTGGG-GG(TG) 1 GG 48 SEQ ID NO:24-(6 bases) GGGTGG-GG(GT) 1 GG 60 SEQ ID NO:25-(6 bases) TTGTTT-TT(GT) 1 TT 34 SEQ ID NO:26-(6 bases) AAGTAA-AA(GT) 1 AA 13 SEQ ID NO:27-(6 bases) CCGTCC-CC(GT) 1 CC 11 SEQ ID NO:28-(6 bases) TGGTTG-TG(GT) 1 TG 42
  • GT SEQ ID NO:25 inhibited proliferation 60% and AGT SEQ ID NO:49 inhibited proliferation 45%.
  • Comparison of the relative potency of GT SEQ ID NO:25 and AGT SEQ ID NO:49 using PHARM/PCS-4 Software showed the potency of GT SEQ ID NO:25 to be 3.4 times that of AGT SEQ ID NO:49.
  • AGT SEQ ID NO:49-phosphorothioate is reported to inhibit telomerase activity and to induce apoptosis in Burkitt lymphoma cells (Mata et al. Toxicol. Appl. Pharmacol. 144:189, 1997).
  • telomerase activity To determine telomerase activity, extracts from 2 ⁇ 10 5 Jurkat T cells were assayed using the PCR-telomeric repeat amplification protocol (TRAP) (Roche, Laval, Québec, Canada). At concentrations between 1 and 100 ⁇ g/ml, GT SEQ ID NO:25-phosphodiester showed between 0 and 10% anti-telomerase activity, whereas AGT SEQ ID NO:49-phosphorothioate showed between 30 and 75% anti-telomerase activity. Neither GT SEQ ID NO:25-phosphorothioate nor GT SEQ ID NO:49-phosphodiester showed any anti-telomerase actvity.
  • TRIP PCR-telomeric repeat amplification protocol
  • HL-60 promyelocytic leukemia cells are a p53 mutated human suspension tumor model. HL-60 cells were incubated with 6 base GT sequences (Table 6). TABLE 6 % inhibition of HL-60 human promyelocytic leukemia cell proliferation SEQUENCE % INHIBITION TGTGTG-(TG) 3 7 SEQ ID NO:9-(6 bases) GTGTGT-(GT) 3 13 SEQ ID NO:10-(6 bases) TTTGTT-TT(TG) 1 TT 13 SEQ ID NO:23-(6 bases) GGTGGG-GG(TG) 1 GG 18 SEQ ID NO:24-(6 bases) GGGTGG-GG(GT) 1 GG 35 SEQ ID NO:25-(6 bases) TTGTTT-TT(GT) 1 TT 16 SEQ ID NO:26-(6 bases) As shown in Table 6, 6 base GT sequences inhibited HL-60 cell proliferation.
  • MCF-7 human breast cancer cells are a caspase 3 negative, estrogen-dependent human solid tumor model.
  • MCF-7 cells (5 ⁇ 10 5 cells/ml) were incubated with 3 and 6 base GT sequences (Table 7).
  • TABLE 7 % inhibition of MCF-7 human breast cancer cell proliferation SEQUENCE % INHIBITION TGT-(TG)T ⁇ 6 SEQ ID NO:7-(3 bases) GTG-(GT)G 18 SEQ ID NO:8-(3 bases) TGTGTG-(TG) 3 6 SEQ ID NO:9-(6 bases) GTGTGT-(GT) 3 31 SEQ ID NO:10-(6 bases) TTTGTT-TT(TG) 1 TT 7 SEQ ID NO:23-(6 bases) GGTGGG-GG(TG) 1 GG 41 SEQ ID NO:24-(6 bases) GGGTGG-GG(GT) 1 GG 41 SEQ ID NO:25-(6 bases) TTGTTT-TT(GT) 1 TT 20 SEQ ID NO:26-
  • PC-3 prostate cancer cells are a p53 mutated, androgen-independent human solid tumor model.
  • PC-3 cells (5 ⁇ 10 5 cells/ml) were incubated with 3 and 6 base GT sequences (Table 8).
  • TABLE 8 % inhibition of PC-3 human prostate cancer cell proliferation SEQUENCE % INHIBITION TGT-(TG)T 8 SEQ ID NO:7-(3 bases) GTG-(GT)G 13 SEQ ID NO:8-(3 bases) TGTGTG(TG) 3 16 SEQ ID NO:9-(6 bases) GTGTGT-(GT) 3 37
  • SEQ ID NO:24-(6 bases) GGGTGG-GG(GT) 1 GG 38 SEQ ID NO:25-(6 bases) TTGTTT-TT(GT) 1 TT 18 SEQ ID NO:26
  • LNCaP prostate cancer cells are a TGF-beta 1 receptor negative, androgen-independent, metastatic human solid tumor model.
  • LNCaP cells (5 ⁇ 10 5 cells/ml) were incubated with 6 base GT sequences (Table 9).
  • TABLE 9 % inhibition of LNCaP human prostate cancer cell proliferation SEQUENCE % INHIBITION TGTGTG-(TG) 3 ⁇ 9 SEQ ID NO:9-(6 bases) GTGTGT-(GT) 3 ⁇ 4 SEQ ID NO:10-(6 bases) TTTGTT-TT(TG)TT 14 SEQ ID NO:23-(6 bases) GGTGGG-GG(TG)GG 17 SEQ ID NO:24-(6 bases) GGGTGG-GG(GT)GG 18 SEQ ID NO:25-(6 bases) TTGTTT-TT(GT)TT 22 SEQ ID NO:26-(6 bases) As shown in Table 9, 6 base GT sequences inhibited LNCaP cell proliferation.
  • THP-1 acute monocytic leukemia cells are a p53 null human suspension tumor model.
  • THP-1 cells (1.6 ⁇ 10 6 cells/ml) were incubated with 6 base GT sequences (Table 10).
  • TABLE 10 % inhibition of THP-1 human acute monocytic leukemia cell proliferation SEQUENCE % INHIBITION TGTGTG-(TG) 3 0 SEQ ID NO:9-(6 bases) GTGTGT-(GT) 3 11 SEQ ID NO:10-(6 bases) TTTGTT-TT(TG) 1 TT 8 SEQ ID NO:23-(6 bases) GGTGGG-GG(TG) 1 GG 6 SEQ ID NO:24-(6 bases) GGGTGG-GG(GT) 1 GG 15 SEQ ID NO:25-(6 bases) TTGTTT-TT(GT) 1 TT 1 SEQ ID NO:26-(6 bases) As shown in Table 10, 6 base GT sequences inhibited THP-1 cell proliferation.
  • OVCAR-3 ovarian cancer cells are a p53 mutated, p21/waf-1/Cip deleted, metastatic human solid tumor model.
  • OVCAR-3 cells (5 ⁇ 10 5 cells/ml) were incubated with 2, 6 and 18 base GT sequences and with a 6 base AGT sequence (Table 11).
  • SK-OV-3 ovarian cancer cells are a p53 mutated, p21/waf-1/Cip deleted, p15 ink4B deleted, p16 Ink4 deleted, metastatichuman solid tumor model.
  • SK-OV-3 cells (5 ⁇ 10 5 cells/ml) were incubated with 2, 6 and 18 base GT sequences (Table 12).
  • Jurkat human leukemia T cells (Table 13), LNCaP human prostate cancer cells (5 ⁇ 10 5 cells/ml) (Table 14) and MCF-7 human breast cancer cells (5 ⁇ 10 5 cells/ml) (Table 15) were incubated with 6 base GT sequences, having either oxygen (phosphodiester) or sulfur (phosphorothioate) as a nonbridging atom on the phosphate group.
  • Jurkat human leukemia T cells (Table 16) and MCF-7 human breast cancer cells (5 ⁇ 10 5 cells/ml) (Table 17) were incubated with the 6 base GT SEQ ID NO:25, wherein either oxygen (phosphodiester) or sulfur (phosphorothioate) was the nonbridging atom on the phosphate group.
  • EL-4 murine lymphoma T cells are a suspension tumor model. EL-4 murine lymphoma T cells were incubated with 6, 18, 27 and 33 base GT sequences and with a 15 base ACG sequence (Table 18). TABLE 18 % inhibition of EL-4 murine T lymphoma cell proliferation SEQUENCE % INHIBITION GGGTGG-GG(GT) 1 GG 4 SEQ ID NO:25-(6 bases) GGGTGG-GG(GT) 1 GG ⁇ 8 SEQ ID NO:25-(6 bases phosphorothioate) GTGTGTGTGTGTGTGTGTGTGTGTGTGTGTGTGTGTGTGTGTGTGTGTG-(G 1 T) 13 G 1 SEQ ID NO:1-(27 bases) GTGTGTTTGGTGGTTTTGTTTGTTGTTTTTTTG ⁇ 1 SEQ ID NO:66-(33 bases) AACCACAAGCCCAAC ⁇ 6 SEQ ID NO:67-(15 bases) GTGTGT-(GT) 3 ⁇ 2 SEQ ID NO:
  • A20 murine leukemia B cells are a suspension tumor model.
  • A20 murine leukemia B cells were incubated with 6 base GT sequences (Table 19).
  • TABLE 19 % inhibition of A20 murine B leukemia cell proliferation SEQUENCE % INHIBITION TGTGTG-(TG) 3 22 SEQ ID NO:9-(6 bases) GTGTGT-(GT) 3 9 SEQ ID NO:10-(6 bases) TTTGTT-TT(TG) 1 TT 5 SEQ ID NO:23-(6 bases) GGTGGG-GG(TG) 1 GG 9 SEQ ID NO:24-(6 bases) GGGTGG-GG(GT) 1 GG 11 SEQ ID NO:25-(6 bases) TTGTTT-TT(GT) 1 TT 15 SEQ ID NO:26-(6 bases) As shown in Table 19, 6 base GT sequences inhibited proliferation of A20 murine B leukemia cells.
  • D-17 canine osteosarcoma cells and CF-51 canine mammary gland cancer cells are solid tumor models.
  • D-17 canine osteosarcoma cells (5 ⁇ 10 5 cells/ml) (Table 20) and CF-51 canine mammary gland cancer cells (5 ⁇ 10 5 cells/ml) (Table 21) were incubated with 6, 9, 17, 27 and 33 base GT-sequences and with a 15 base ACG sequence.
  • Jurkat human leukemia T cells were incubated with suboptimal concentrations (5.0 ⁇ g/ml) of 6 base GT sequences (Table 23). TABLE 23 % inhibition of Jurkat human leukemia T cell proliferation SEQUENCE % INHIBITION GGGTGG-GG(GT) 1 GG 5 SEQ ID NO:25-(6 bases) TTGTTT-TT(GT) 1 GG ⁇ 2 SEQ ID NO:26-(6 bases) GG(GT) 1 GG + TT(GT) 1 GG 14 SEQ ID NO:25 + SEQ ID NO:26 TGGTTG-TG(GT) 1 TG ⁇ 1 SEQ ID NO:29-(6 bases) TGGTTG-TG(GT) 1 TG 2 SEQ ID NO:10-(6 bases) TG(GT) 1 TG + TG(GT) 1 TG 9 SEQ ID NO:29 + SEQ ID NO:10 GGTTGG-GG(TT) 1 GG 4 SEQ ID NO:41-(6 bases) TTGTGG-TT
  • Jurkat human leukemia T cells were incubated with 1.0 ⁇ g/ml of 6 base GT SEQ ID NO:25 and of 27 base GT SEQ ID NO:1 in the presence of 0, 0.1, 1.0 or 10.0 ⁇ g/ml of 5-fluorouracil or cisplatin (Table 24).
  • 5-fluorouracil is an antimetabolite that interferes with DNA and RNA synthesis.
  • Cisplatin. is an alkylating agent that cross-links DNA and inhibits DNA precursors.
  • GT SEQ ID NO:25 As shown in Table 24, 6 base GT SEQ ID NO:25 and 27 base GT SEQ ID NO:1 potentiated the antineoplastic effect of 0.1 and 1.0 ⁇ g/ml of 5-fluorouracil on Jurkat T cell proliferation and GT SEQ ID NO:25 potentiated the antineoplastic effect of 0.1 and 1.0 ⁇ g/ml cisplatin on Jurkat T cell proliferation.
  • MCF-7 human breast cancer cells (5 ⁇ 10 5 cells/ml) were incubated with 1.0 ⁇ g/ml of 6 base GT SEQ ID NO:25 and of 27 base GT SEQ ID NO:1 in the presence of 0, 0.1, 1.0 or 10.0 ⁇ g/ml of 5-fluorouracil or tamoxifen (Table 25).
  • Tamoxifen is an estrogen antagonist.
  • the melting temperature (Tm) of 6 base GT SEQ ID NO:25 was 2.5° C. and increased to 56.8° C. with GT SEQ ID NO:68, to 76.3° C. with GT SEQ ID NO:69 and to 86.3° C. with GT SEQ ID NO:70.
  • BCG derived sequences are reported to inhibit tumor growth in vivo (Kataoka et al. Jpn. J. Cancer Res. 83:244, 1992).
  • A-2 SEQ ID NO:72
  • BCG A-4 SEQ ID NO:74
  • Cell cycle stage was determined using a CYCLETESTM PLUS DNA commercial kit (Becton Dickinson). Briefly, nuclei from cells were obtained by dissolving the cell membrane in a nonionic detergent, eliminating the cell cytoskeleton and nuclear proteins with trypsin, digesting the cellular RNA with RNase and stabilizing the nuclear chromatin with spermine. Propidium iodide was added to the cell nuclei and their fluorescence was analyzed in a flow cytometer equipped with electronic doublet discrimination capability (FACSCalibur, Becton Dickinson, San Jose, Calif.). Accumulation of cells in G 0 /G1, early S (SE), mid S (SM), late S (SL) or G 2 /M phases of the cell cycle was analyzed using MODFIT LT software (Verity Software House Inc., Topsham, Mass.).
  • phosphatidylserine in the plasma membrane during apoptosis was measured by flow cytometry using FITC-conjugated annexin V (BD Pharmingen, San Diego, Calif.). NuMA release into the supernatant was determined using a commercial ELISA kit (Oncogen/Calbiochem, Cambridge, Mass.).
  • Jurkat human leukemia T cells were incubated with 50 ⁇ M of 3, 4, 5, 6 and 7 GT base sequences, a 5 base ACGT sequence, 6 base AG, GG, AGT and CGT sequences and a 7 base GG sequence.
  • Table 31 shows % of cells in apoptosis (positive for phosphatidyl-serine/annexin V staining (PS/A-V)) and % NuMA released from the cells.
  • Jurkat human leukemia T cells were incubated for 24 h with 6 base GT SEQ. NO:25. Cells were collected by centrifugation, suspended in PBS containing 1% FBS and loaded with 10 ⁇ M Fluo-3-AM for 1 h at 37° C. Cell fluorescence was measured at 488 nm excitation and 530 nm emission (FL1 detector). Data were analyzed on a FACSCALIBUR using the program CellQUEST (Becton Dickinson).
  • Apoptosis can be initiated by ligands that bind to cell surface receptors including, but not limited to, Fas (CD95) and tumor necrosis factor (TNF). Fas binding to Fas Ligand and TNF binding to TNF Receptor 1 initiate intracellular signaling resulting in the activation of cysteine aspartyl proteases (caspases). Caspases initiate the lethal proteolytic cascade of apoptosis execution associated with nuclear DNA-fragmentation, release of nuclear matrix proteins (NuMA) and loss of cell substrate contact.
  • Fas CD95
  • TNF Receptor 1 tumor necrosis factor
  • Jurkat human leukemia T cells were incubated with GT-rich or AC-rich sequences derived from the murA gene of Mycobacterium phlei (GenBank: Accession Number X99776). Inhibition of proliferation was measured by the reduction of MTT as in Example 3, cell cycle arrest was detected by flow cytometry using propidium iodide as in Example 21 and apoptosis was evaluated by flow cytometry using annexin-V-FITC as in Example 23.
  • Caspases recognize 3 major peptide substrate sequences: 1) Tyr-Val-Ala-Asp (YVAD, caspase-1, -4 and -5) (SEQ ID NO:84); 2) Asp-Glu-Val-Asp (DEVD, caspase-2, -3 and -7) (SEQ ID NO:85); and, 3) Ile-(Leu)-Glu-X-Asp (I(L)EXD; caspase-8 and -10) (SEQ ID NO:86) (Thomberry et al. J. Biol. Chem. 272:17907, 1997).
  • Sequence recognition in a protein target results in a limited and specific proteolysis of the target as, in a first example, the modulation of caspase 7 activation by caspase 3 or, as in a second example, the degradation of structural protein targets including, but not limited, to lamins or, as in a third example, the activation of enzymes including, but not limited to, PARP.
  • NH 2 —XXXD-COO-GT sequence constructs are generated by chemical conjugation of a chemically protected GT sequence or of a chemically protected AC sequence to a chemically protected peptide selected from the group consisting of NH 2 —YVAD-COOH (SEQ ID NO:84), NH 2 -DEVD-COOH (SEQ ID NO:85) and NH 2 -IEGD-COOH(SEQ ID NO:87) using an oligonucleotide synthesized with a 5′-C 2 amide spacer arm and standard amide-carboxyl water soluble carbohexiimide conjugation techniques (Guy et al. J. Chromatography. B. Biomed. Sci. Appl. 706:149, 1998). Reactive carboxylate and reactive amine groups are deprotected subsequent to conjugation,
  • PGT sequence constructs including, but not limited to, NH 2 —YVAD-COO-GT; NH 2 -DEVD-COO-GT; and, NH 2 —I(L)EXD-COO-GT are cleaved at the carboxylate function between D and the GT sequence by enzymes including, but not limited to, caspases, cancer metastasisassociated enzymes, collagenase and metalloproteinases. Such cleavage results in the release of the caspase-activating GT sequence from the PGT.
  • the resulting increase in intracellular caspase activity can, for example, enhance the therapeutic effect of chemotherapeutic agents in multidrug resistant cancer cells or the immune response to weakly antigenic stimuli.
  • caspase activation is determined colorimetrically using an assay based on the cleavage of a caspase-specific peptide conjugated to the color reporter molecule p-nitroanilide, which can be quantitated spectrophotometrically at a wavelength of 405 nm.
  • Jurkat T cell leukemia cells were incubated for 72 h with 33 base GT SEQ ID NO:66; 11 base GT SEQ ID NO:81 (bases 1-11 of GT SEQ ID NO:66), 11 base GT SEQ ID NO:82 (bases 12-22 of GT SEQ ID NO:66), 11 base GT SEQ ID NO:83 (bases 23-33 of GT SEQ ID NO:66) and 15 base ACG SEQ ID NO:67.
  • Active caspase 3 (17-22 kDa) was determined using FITC conjugated antibody (Clone: C92-605) as in Example 28.
  • Caspase 3 activation also was determined colorimetrically as in Example 28. As As shown in FIG. 2B , caspase 3 activity in 33 base GT SEQ ID NO:66 and 11 base GT SEQ ID NOs:81, 82 and 83 treated cells was 105%, 77%, 100% and 60% greater than that in control cells, whereas in 15 base ACG SEQ ID NO:67 treated cells caspase 3 activation was approximately the same as in control cells.
  • Jurkat T cell leukemia cells were incubated for 72 h with 6 base GT SEQ ID NO:25.
  • the cells were washed 3 ⁇ with PBS, lysed with 10 mM HEPES, pH 7.5 containing 5 mM MgCl 2 , 1 mM dithiothreitol, 1.5 nM aprotinin, 10 mM leupeptin and 2.5 ⁇ m Na orthovanadate, and the protein content of the lysate was determined (Bradford J. Anal. Biochem. 72:248, 1976).
  • Activated caspase 7 and PARP cleavage were detected by Western blot analysis.
  • Lysate was mixed with Laemmli buffer (Laemmli U. Nature 15:680, 1970), shaken, and heated at 100° C. for 4 min.
  • Fifty ⁇ g of protein was added to each lane and the proteins were separated by electrophoresis in a 10% (caspase) or a 17% (PARP) sodium dodecyl sulfate-polyacrylamide gels (SDS-PAGE) at a constant voltage of 100 V for about 1.5 h.
  • the separated proteins were electroblotted onto a PVDF membrane. Equal protein loading was monitored by Ponceau red staining of the membrane.
  • the membrane was blocked overnight at 4° C. with a buffer containing 1% Tris-buffered saline (2 mM Tris-HCl, 13.7 mM NaCl, pH 7.6, and 0.1% polyethylenesorbitan monolaurate (TWEEN 20) (TBST) +5% non-fat dry milk.
  • TBST polyethylenesorbitan monolaurate
  • the membrane was washed and was incubated for 1 h at RT with a mouse monoclonal IgG anti-caspase 7 antibody (Pharmingen) (diluted 1:1000 in TBST+1% BSA) or with a mouse monoclonal IgG anti-PARP antibody (diluted 1:1000 in TBST+1% BSA) (Pharmingen).
  • IgG bound to caspase 7 or to PARP was detected with sheep anti-mouse IgG conjugated to horseradish peroxidase (diluted 1:1000 in TBST+5% non-fat dry milk) (Pharmingen). Blots were developed using an enhanced chemiluminescence detection system (ECL, Amersham, Corp., Amersham, UK).
  • 6 base GT SEQ ID NO:25, induced processing of inactive pro-caspase 7 (30 kDa) to active caspase 7 (19-20 kDa) and active PARP to its inactive 85 kDa degradation product.
  • Jurkat human leukemia T cells are incubated for 72 h with NH 2 —YVAD-CO-GG(GT)GG, NH 2 -DEVD-CO-GG(GT)GG, NH 2 —EGD-COO-GG(GT)GG, NH 2 -YVAD-CO-AACCACAAGCCCAAC, NH 2 -DVED-CO-AACCACAAGCCCAAC and NH 2 —IEGD-CO-AACCACAAGCCCAAC. Caspase 3 and caspase 7 activation are determined.
  • NH 2 —YVAD-CO-GT, NH 2 -DEVD-CO-GT and NH 2 —IEGD-COO-GT each induce processing of inactive caspase 3 to active caspase 3 and of inactive caspase 7 to active caspase 7, whereas NH 2 —YVAD-CO-ACG, NH 2 -DVED-CO-ACG and NH 2 —IEGD-CO-ACG do not induce processing of inactive pro-caspase 3 to active caspase 3 or of inactive caspase 7 to active caspase 7.
  • caspase activity within caspase containing cells mediates the release of caspase activating GT sequences from NH 2 —XXXD-COO-GT constructs by proteolysis/hydrolysis. This results in positive amplification of caspase activity (increased levels of caspase 3 and caspase 7) within the cells by the released caspase-activating GT sequences.
  • THP-1 human acute monocytic leukemia cells were incubated with 2, 3, 6, 9, 12, 14, 15 and 18 base GT sequences and production of the cyotkine IL-6 was determined (Table 35).
  • Table 35 Cytokine production by THP-1 human acute monocytic leukemia cells SEQUENCE ⁇ IL-6 TG-(TG) 1 T 2.7x SEQ ID NO:7-(3 bases) TG-(TG) 1 2.9x SEQ ID NO:51-(2 bases) TGTGTG-(TG) 3 4.0x SEQ ID NO:9-(6 bases) GTGTGT-(GT) 3 5.0x SEQ ID NO:10-(6 bases) TGTGTGTG-(TG) 4 T 5.4x SEQ ID NO:11-(9 bases) GTGTGTGTG-(GT) 4 G 5.4x SEQ ID NO:12-(9 bases) TGTGTGTGTGT-(TG) 6 5.7x SEQ ID NO:13-(12 bases) GTGTGTGTGTGT-(GT
  • THP-1 human acute monocytic leukemia cells were incubated with 6 base GT, AG, CG, GG, AGT and CGT sequences and production of the cytokines IL-12 and IL-6 was determined (Table 36).
  • Cytokine production by THP-1 human acute monocytic leukemia cells SEQUENCE ⁇ IL-12 ⁇ IL-6 TGTGTG-(TG) 3 SEQ ID NO:9 1.8x 4.0x GTGTGT-(GT) 3 SEQ ID NO:10 2.2x 5.0x TTTGTT-TT(TG) 1 TT SEQ ID NO:23 3.5x 4.9x GGTGGG-GG(TG) 1 GG SEQ ID NO:24 3.7x 6.9x GGGTGG-GG(GT) 1 GG SEQ ID NO:25 2.3x 3.1x TTGTTT-TT(GT) 1 TT SEQ ID NO:26 3.5x 5.3 AAGTAA-AA(GT) 1 AA SEQ ID NO:27 6.0x 12.8x CC
  • Table 37 summarizes the induction of IL12 and IL-6 synthesis by 6 base sequences. TABLE 37 IL-6 and IL-12 synthesis induced by 6 base sequences Fold IL-12 synthesis IL-6 synthesis increase SEQ ID NOs: SEQ ID NOs: ⁇ 2.0 9, 31, 32, 33, 34, 35, 36, 38, 39, 40, 44, 45, 49 >2.0 and 10, 23, 24, 25, 26, 27, 28, 29, 9, 10, 23, 24, 25, 26, 30, 31, ⁇ 10.0 30, 37, 40, 41, 42, 43, 44, 45 32, 33, 38, 39, 46, 49 >10.0 25, 27, 29, 34, 35, 36, 37, 40, 41, 42, 43, 44, 45
  • BCG derived sequences A-3 (SEQ ID NO:73), A-4 (SEQ ID NO:74), A-6 (SEQ ID NO:75), A-7 (SEQ ID NO:76), M3 (SEQ ID NO:77) and Alpha 1 (SEQ ID NO:78) are reported to activate NK cells in vivo (Kataoka et al. Jpn. J. Cancer Res. 83:244, 1992).
  • THP-1 human acute monocytic leukemia cells were incubated with 45 base BCG-derived sequences and production of the cytokines IL-12 and IL-6 was determined (Table 38).
  • THP-1 human acute monocytic leukemia cells were incubated with 6 base GT sequences, having either an oxygen (phosphodiester) or a sulfur (phosphorothioate) as the nonbridging atom on the phosphate groups and production of the cytokine IL-12 was determined (Table 39).
  • THP-1 human acute monocytic leukemia cells were incubated with 6 base GT SEQ ID NO:25, having either an oxygen (phosphodiester) or a sulfur (phosphorothioate) as the nonbridging atom on the phosphate groups and production of the cytokine IL-12 was determined (Table 40).
  • PBMCs Peripheral blood mononuclear cells
  • 6 base GT, AGT, CGT, AG, CG and GG sequences increased human PBMC cell production of the cytokines IL1beta, IL-6, IL-10 and IL-12.
  • PBMCs were isolated from 4 healthy chimpanzees as in Example 35. Chimpanzee PBMCs were incubated with 6 base GT, AGT, CGT, AG, CG and GG sequences and production of the cytokines IL-10, IL-12 and TNF-alpha was determined (Table 41).
  • PBMCs were isolated from 4 healthy rhesus monkeys as in Example 35. PBMCs were incubated with 6 base GT, AGT, CGT, AG, CG ands GG sequences and production of the cytokines IL-6, IL-12 and TNF-alpha was determined (Table 42).
  • MCF-7 human breast cancer cells are implanted subcutaneously as xenografts, in female nude BALB/c mice.
  • the mice are divided into 6 groups of 10 mice.
  • group 1 mice receive saline
  • group 2 mice receive 6 base GT SEQ ID NO:25
  • group 3 mice receive receive 5-fluorouracil
  • group 4 mice receive tamoxifen
  • group 5 mice receive 6 base GT SEQ ID NO:25+5-fluorouracil
  • group 6 mice receive 6 base GT SEQ ID NO:25+tamoxifen.
  • the mice are sacrificed and tumor mass is determined.
  • Group 1 mice have the most tumor mass
  • groups 2, 3 and 4 mice have less tumor mass than group 1 mice
  • groups 5 and 6 mice have less tumor mass than groups 1, 2, 3 and 4 mice.
  • LNCaP human prostate cancer cells are implanted subcutaneously, as xenografts, in male nude nu/nu mice.
  • the mice are divided into 5 groups of 10 mice.
  • group 1 mice receive saline
  • group 2 mice receive 3 base SEQ ID NO:8
  • group 3 mice receive 6 base GT SEQ ID NO:25
  • group 4 mice receive 6 base AG
  • SEQ ID NO:45 and group 5 mice receive 45 base BCG A-3 SEQ ID NO:69.
  • the mice are sacrificed and timor mass is determined.
  • Group 1 mice have the most tumor mass
  • group 5 mice have less tumor mass than group 1 mice and groups 2
  • 3 and 4 mice have less tumor mass than groups 1 and 5 mice.
  • EL-4 murine T lymphoma cells are implanted into C57/BL6 mice.
  • the mice are divided into 6 groups of 10 mice.
  • group 1 mice receive saline
  • group 2 mice receive 3 base GT SEQ ID NO:8
  • group 3 mice receive 6 base SEQ ID NO:25
  • group 4 mice receive 6 base AG SEQ ID NO:45
  • group 5 mice receive 18 base GT SEQ ID NO: 18
  • group 6 mice receive 27 base GT SEQ ID NO:1.
  • the mice are sacrificed and tumor mass is determined.
  • Group 1 mice have the most tumor mass
  • groups 2, 3, 4, 5 and 6 mice have less tumor mass than group 1 mice.
  • Human colon cancer cell lines are maintained as adherent cell cultures.
  • Cells in the exponential growth phase are treated with 2-20 base GT, GA, GC or GG sequences in the dose range 0 ⁇ g/ml to 100 ⁇ l/ml for 24-72 hours.
  • Inhibition of cell proliferation is measured by MTT reduction, cell cycle arrest by flow cytometry and apoptosis by annexin-V binding or NuMA release.
  • GT, GA, GC or GG sequences inhibit proliferation, induce cell cycle arrest and induce apoptosis in the colon cancer cell lines.
  • SCID mice bearing subcutaneous human colorectal cancer cell lines are treated with saline or with 2-20 base GT, GA, GC or GG sequences, having anti cancer activity against human colorectal cancer cell lines in vitro-
  • Mice treated with 2-20 base GT, GA, GC or GG sequences, having anti-cancer activity against human colorectal cancer cell lines in vitro show a significant reduction in tumor mass compared with mice treated with saline.

Abstract

The present invention provides a composition and method comprising a 2-20 base 3′-OH, 5′-OH synthetic oligonucleotide (sequence) selected from the group consisting of (GxTy)n, (TyGx)n, a(GxTy)n, a(TyGx)n, (GxTy)nb, (TyGx)nb, a(GxTy)nb, a(TyGx)nb, wherein x and y is an integer between 1 and 7, n is an integer between 1 and 12, a and b are one or more As, Cs, Gs or Ts aid wherein the sequence induces a response selected from the group consisting of induction of cell cycle arrest, inhibition of proliferation, activation of caspases and induction of apoptosis in cancer cells and production of cytokines by immune system cells.

Description

  • This patent application claims priority to U.S. provisional patent application Ser. No. 60/170,325, filed Dec. 13, 1999 and U.S. provisional patent application Ser. No. 60/228,925, filed Aug. 29, 2000.
  • FIELD OF THE INVENTION
  • The present invention relates to an oligonucleotide composition for the treatment of cancer.
  • BACKGROUND OF THE INVENTION
  • Cancer is an aberrant net accumulation of atypical cells, which can result from an excess of proliferation, an insufficiency of cell death, or a combination of the two.
  • Proliferation is the culmination of a cell's progression through the cell cycle resulting in the division of one cell into two cells. The 5 major phases of the cell cycle are G0, G1, S, G2 and M. During the G0 phase, cells are quiescent. Most cells in the body, at any one time, are in this stage. During the G1 phase, cells, responding to signals to divide, produce the RNA and the proteins necessary for DNA synthesis. During the S-phase (SE, early S-phase; SM, middle S-phase; and SL, late S-phase) the cells replicate their DNA. During the G2 phase, proteins are elaborated in preparation for cell division. During the mitotic (M) phase, the cell divides into two daughter cells. Alterations in cell cycle progression occur in all cancers and may result from over-expression of genes, mutation of regulatory genes, or abrogation of DNA damage checkpoints (Hochhauser D. Anti-Cancer Chemotherapeutic Agents 8:903, 1997).
  • Unlike cancer cells, most normal cells cannot proliferate indefinitely due to a process termed cellular senescence. Cellular senescence is a programmed cell death response leading to growth arrest of cells (Dimri et al. Proc. Natl. Acad. Sci. USA 92:20, 1995). DNA damage, exposure of colon, breast and ovarian cancer cells to toposiomerase inhibitors and exposure of nasopharyngeal cancer cells to cisplatin are reported to prevent proliferation of these cells by induction of senescence (Wang et al. Cancer Res. 58:5019, 1998; Poele et al. Br. J. Cancer 80:9, 1999).
  • Synthetic oligonucleotides are polyanionic sequences that are internalized in cells (Vlassov et al. Biochim. Biophys. Acta 1197:95, 1994). Synthetic oligonucleotides are reported that bind selectively to nucleic acids (Wagner, R. Nature: 372:333, 1994), to specific cellular proteins (Bates et al. J. Biol. Chem. 274:26369, 1999) and to specific nuclear proteins Scaggiante et al. Eur. J. Biochem. 252:207, 1998) to inhibit proliferation of cancer cells.
  • Synthetic 27 base sequences containing guanine (G) and variable amounts of thymine (T) (oligonucleotide GTn), wherein n is ≧1 or ≦7 and wherein the number of bases is ≧20 (Scaggiante et al. Eur. J. Biochem. 252:207, 1998), are reported to inhibit growth of cancer cell lines by sequence specific binding to a 45 kDa nuclear protein, whereas GTn, wherein the number of bases is ≦20, are reported to be inactive against cancer cell lines (Morassutti et al. Nucleosides and Nucleotides 18:1711, 1999). Two synthetic GT-rich oligonucleotides of 15 and 29 bases with 3′ aminoalkyl modifications are reported to form G-quartets that bind to nucleolin and to inhibit proliferation of cancer cell lines (Bates et al. J. Biol. Chem. 274:26369, 1999). The synthetic 6 base TTAGGG-phosphorothioate, having a sequence identical to that of the mammalian telomere repeat sequence, is reported to inhibit proliferation of Burkitt's lymphoma cells in vitro and in vivo (Mata et al. Toxicol. Applied Pharmacol. 144:189, 1997). However, the synthetic 6 base TTAGGG-phosphodiester is reported to have no antitelomerase activity (U.S. Pat. No. 5,643,890).
  • Cell death is effected by immune-mediators that promote apoptosis, and by apoptosis inducers that directly initiate pathways leading to cell death (Muzio et al. Cell 85:817, 1996; Levine, A. Cell 88:323, 1997). Apoptosis is an active cellular death process characterized by distinctive morphological changes that include condensation of nuclear chromatin, cell shrinkage, nuclear disintegration, plasma membrane blebbing, and the formation of membrane-bound apoptotic bodies (Wyllie et al. Int. Rev. Cytol. 68:251, 1980). A molecular hallmark of apoptosis is degradation of the cell's nuclear DNA into oligonucleosomal-length fragments as the result of activation of endogenous endonucleases (Wyllie A. Nature 284:555, 1980).
  • Caspases (cysteine-aspartyl-specific proteases) have been implicated as key enzymes in the execution of the late stage of apoptosis. The caspase family consists of at least fourteen related cysteine aspartyl proteases. All the caspases contain a conserved QACXG (where X is R, Q or G) pentapeptide activesite motif (Cohen G. Biochim. Biophys. Acta 1366:139, 1997). A number of caspases are synthesized as inactive proenzymes that are activated following cleavage at caspase specific cleavage sites (Cohen G. Biochim. Biophys. Acta 1366:139, 1997) or as inactive enzymes that require association with regulatory molecules for activation (Stennicke et al. J. Biol. Chem. 274:8359, 1999).
  • In addition to their role in apoptosis, caspases are involved in activation and proliferation of B and T lymphocytes, in cytokine maturation during inflammation, in differentiation of progenitor cells during erythropoiesis and in development of lens fiber (Fadeel et al. Leukemia 14:1514, 2000). With respect to B and T lymphocytes, caspase 3 is processed during activation of B lymphocytes and of CD4 (+), CD8 (+), CD45RA(+) and CD45RO (+) subsets of T lymphocytes (Alam et al. J. Exp. Med. 190:1879, 1999). Moreover, stimulation of T lymphocytes by mitogens and by interleukin-2 is associated with activation of the caspase pathway and with cleavage of PARP (Wilheim et al. Eur. J. Immunol. 28:891, 1998). With respect to cytokines, caspase 3 activity is necessary for the release of IL-2 by activated T lymphocytes (Posmantur et al. Exp. Cell Res. 244:302, 1998) and for the processing and maturation of the pro-inflammatory cytokine interleukin-16 (Zhang et al. J. Biol. Chem. 273:1144, 1998). With respect to erythropoiesis, caspase activation is involved in erythropoiesis regulation and has been shown to modulate GATA-1, a nuclear regulatory protein crucial for the maturation of erythroid precursors (De Maria, et al. Nature 401:489, 1999).
  • Cytolysis is the complete or partial destruction of a cell and is mediated by the immune system. Activated macrophages and monocytes produce bioactive molecules that include, but are not limited to cytokines. Cytokines, include, but are not limited to, interleukin (IL)-1, IL-1 beta, IL-6, IL-10, IL-12, and TNF-alpha.
  • IL-1beta reduces bone marrow cell sensitivity to cytoreductive drugs, to radiation and to in vitro tumor cell purging with drugs in autologous bone marrow transplantation (Dalmau et al. Bone Marrow Transplant. 12:551, 1993).
  • IL-6 induces B cell differentiation, stimulates IgG secretion (Taga et al. J. Exp. Med. 166:967, 1987), induces cytotoxic T cell differentiation (Lee et al. Vaccine 17:490, 1999), promotes megakaryocyte maturation (Ishibashi et al. Proc. Natl. Acad. Sci. USA 86:8953, 1989) and functions both as an anti proliferative factor (Mori et al. Biochem. Biophys. Res. Comm. 257:609, 1999; Alexandroff et al. Biochem. Soc. Trans. 25:270, 1997; Takizawa et al. Cancer Res. 53:18, 1993: Novick et al. Cytokine 4:6, 1992) and as a pro-proliferative factor (Okamoto et al. Cancer Res. 57:141, 1997; Okamoto et al. Int. J. Cancer 72:149, 1997; Chiu et al. Clin. Cancer Res. 2:215, 1996; Lu et al. Clin. Cancer Res. 2:1417, 1996) for cancer cells.
  • IL-10 enhances the effectiveness of vaccines in murine tumor models (Kauffman et al. J. Immunother. 22: 489, 1999) and up-regulates anti-cancer autoreactive T cell responses (Alleva et al. Immunobiol. 192:155, 1995).
  • IL-12, alone and in combination with other cytokines, promotes the maturation of leukocytes and induces the secretion of interferon-gamma. IL-12 is reported to have anti-cancer activity (Stine et al. Annals NY Academy of Science 795:420, 1996; Chen et al. Journal of Immunol. 159:351, 1997) including, but not limited to, activation of specific cytolytic T-lymphocytes, activation of natural killer (NK) cells and induction of the anti-angiogenic proteins IP-10 and MiG.
  • TNF-alpha causes necrosis of solid tumors (Porter et al. Trends in Biotech. 9:158, 1991), sensitizes cancer cells to gamma irradiation-induced apoptosis (Kimura et al. Cancer Res. 59:1606, 1999) and protects bone marrow precursor cells from the effects of antineoplastic agents (Dalmau et al. Bone Marrow Transplant. 12:551, 1993).
  • However, most prior art anti-cancer therapies, whether directed to induction of cell cycle arrest, inhibition of proliferation, induction of apoptosis or stimulation of the immune system, have proven to be less than adequate for clinical applications. Many of these therapies are inefficient or toxic, have significant adverse side effects, result in development of drug resistance or immunosensitization, and are debilitating for the recipient
  • Therefore, there is a continuing need for novel compositions and methods that induce cell cycle arrest in cancer cells, inhibit proliferation of cancer cells, activate caspases in cancer cells, induce apoptosis in cancer cells and stimulate cytokine production by immune system cells.
  • SUMMARY OF THE INVENTION
  • The present invention fulfills this need by providing a composition and method comprising a 2 to 20 base 3′-OH, 5′-OH synthetic oligonucleotide (hereinafter “sequence”) selected from the group consisting of (GxTy)n, (TyGx)n, a(GxTy)n, a(TyGx)n, (GxTy)nb, (TyGx)nb, a(GxTy)nb, a(TyGx)nb, wherein x and y is an integer between 1 and 7, n is an integer between 1 and 12, a and b are one or more As, Cs, Gs or Ts, and wherein the sequence induces a response selected from the group consisting of induction of cell cycle arrest, inhibition of proliferation, activation of caspases and induction of apoptosis in cancer cells and production of cytokines by immune system cells.
  • A composition comprising a sequence and a pharmaceutically acceptable carrier is administered to an animal, including a human, having cancer in an amount effective to treat the cancer in the animal. The unexpected and surprising ability of the sequence to induce cell cycle arrest, inhibit proliferation, activate caspases and induce apoptosis and in cancer cells and to stimulate cyotkine production by immune system cells addresses a long felt unfulfilled need in the medical arts and provides an important benefit for animals, including humans.
  • Accordingly, it is an object of the present invention is to provide a composition and method effective to treat a disease in an animal, including a human.
  • Another object of the present invention is to provide a composition and method effective to treat a cancer.
  • Another object of the present invention is to provide a composition and method that induces senescence in cells.
  • Another object of the present invention is to provide a composition and method that induces cell cycle arrest in cells.
  • Another object of the present invention is to provide a composition and method that induces cell cycle arrest in cancer cells.
  • Another object of the present invention is to provide a composition and method that inhibits proliferation of cells.
  • Another object of the present invention is to provide a composition and method that inhibits proliferation of cancer cells.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cells.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of Fas.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of TNFR1.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of p53/p21.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of p21/waf-1/CIP.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of p15ink4B.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of p16ink4.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of caspase 3.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of TGF-beta 1 receptor.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of hormone dependence.
  • Another object of the present invention is to provide a composition and method that induces apoptosis in cancer cells independent of drug resistance.
  • Another object of the present invention is to provide a composition and method that activates caspases in cells.
  • Another object of the present invention is to provide a composition and method that activates caspases in cancer cells.
  • Another object of the present invention is to provide a composition and method to treat an autoimmune disease.
  • Another object of the present invention is to provide a composition and method to treat a lymphoproliferative disease.
  • Another object of the present invention is to provide a composition and method to treat an infection.
  • Another object of the present invention is to provide a composition and method to treat an inflammation.
  • Another object of the present invention is to provide a composition and method to modulate T- or B-cell activation.
  • Another object of the present invention is to provide a composition and method to modulate progenitor cell maturation.
  • Another object of the present invention is to provide a composition and method to modulate erythropoiesis.
  • Another object of the present invention is to provide a composition and method to modulate transcription factors in cells.
  • Another object of the present invention is to provide a composition and method that potentiates the effect of other therapeutic agents on cells.
  • Another object of the present invention is to provide a composition and method that potentiates the effect of chemotherapeutic agents on cancer cells.
  • Another object of the present invention is to provide a composition and method that potentiates the anti-neoplastic effect of radiation.
  • Another object of the present invention is to provide a composition and method that stimulates cytokine production by cells of the immune system.
  • Another object of the present invention is to provide a composition and method that stimulates IL-1 beta production by cells of the immune system.
  • Another object of the present invention is to provide a composition and method that stimulates IL-6 production by cells of the immune system.
  • Another object of the present invention is to provide a composition and method that stimulates IL-10 production by cells of the immune system.
  • Another object of the present invention is to provide a composition and method that stimulates IL-12 production by cells of the immune system.
  • Another object of the present invention is to provide a composition and method that stimulates TNF-α production by cells of the immune system.
  • Another object of the present invention is to provide a composition that is simple to prepare.
  • Another object of the present invention is to provide a composition that is minimally toxic to the recipient.
  • These and other objects, features and advantages of the present invention will become apparent after a review of the following detailed description of the disclosed embodiment and the appended claims.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1. Fluorescence [Fluo-3-AM] of Jurkat human T leukemia cells incubated with 0 μg/ml and 100 μg/ml of 6 base GT SEQ ID NO:25.
  • FIG. 2. Caspase 3 activation in Jurkat human T cell leukemia cells incubated without 0 μg/ml and 100 μg/ml of GT SEQ ID NOs:66, 67, 81, 82 and 83 measured cytometrically (A) and colorimetrically (B).
  • FIG. 3. Caspase activation in Jurkat human T cell leukemia cells. (A) Caspase 3 activation in cells incubated with 0 μg/ml and 100 μg/ml of 6 base GT SEQ ID NO:25; (B) Caspase 7 activation(a) and PARP content (b) in cells incubated with 0 μg/ml and 100 μg/ml of 6 base GT SEQ ID NO:25.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides a composition comprising a 2 to 20 base 3′-OH, 5′OH synthetic oligonucleotide (sequence) selected from the group consisting of (GxTy)n, (TyGx)n, a(GxTy)n, a(TyGx)n, (GxTy)nb, (TyGx)nb, a(GxTy)nb, a(TyGx)nb, wherein x and y is an integer between 1 and 7, n is an integer between 1 and 12, a and b are one or more As, Cs, Gs or Ts, wherein the sequence induces a response selected from the group consisting of induction of cell cycle arrest, inhibition of proliferation, activation of caspases and induction of apoptosis in cancer cells and production of cytokines by immune system cells.
  • A composition comprising a sequence and a pharmaceutically acceptable carrier is administered to an animal, including a human, having cancer in an amount effective to treat the cancer in the animal, including the human. The unexpected and surprising ability of the sequence to induce cell cycle arrest, inhibit proliferation, induce apoptosis and activate caspases in cancer cells and to stimulate cytokine production by immune system cells addresses a long felt unfulfilled need in the medical arts and provides an important benefit for animals, including humans.
  • As used herein the word “sequence” refers to a 2 to 20 base 3′-OH, 5′-OH synthetic oligonucleotide comprising A, C, G and T bases.
  • As used herein, the abbreviations “GT”, “AG” “CG”, “GG”, “AGT” and “CGT” refer to sequences comprising the named bases synthesized in any order.
  • As used herein, the word “response” refers to induction of cell cycle arrest, inhibition of proliferation, activation of caspases and induction of apoptosis, in cancer cells and stimulation of cytokine production by immune system cells.
  • As used herein, the phrases “therapeutic treatment” and “amount effective to” refer to an amount of a sequence effective to induce cell cycle arrest, inhibit proliferation, activate caspases and induce apoptosis in cancer cells and stimulate cytokine production by immune system cells.
  • As used herein, the phrases “suspension tumor model” and “solid tumor models” refer to primary or secondary carcinomas or sarcomas”.
  • As used herein, the phrase “chemotherapeutic” is any agent approved by a regulatory agency of a country or a state government or listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia to treat cancer in an animal, including a human.
  • As used herein, the word “antineoplastic” refers to preventing the development, maturation, proliferation or spread of cancer cells.
  • As used herein, the word “potentiates” refers to a degree of synergism that is greater than the additive activity of each agent.
  • As used herein, the word “synergism” refers to the coordinated action of two or more agents.
  • Administration of an effective amount of a sequence of the present invention to an animal, including a human, is a therapeutic treatment that prevents, treats or eliminates a disease including, but not limited to, cancer, rheumatoid arthritis, lympho-proliferative disorders and asthma. Cancers include, but are not limited to, squamous cell carcinoma, fibrosarcoma, sarcoid carcinoma, melanoma, mammary cancer, lung cancer, colorectal cancer, renal cancer, osteosarcoma, cutaneous melanoma, basal cell carcinoma, pancreatic cancer, bladder cancer, brain cancer, ovarian cancer, prostate cancer, leukemia, lymphoma and metastases derived therefrom.
  • The therapeutic effectiveness of a sequence may be increased by methods including, but not limited to, chemically modifying the base, sugar or phosphate backbone, chemically supplementing or biotechnologically amplifying the sequences using bacterial plasmids containing the appropriate sequences, complexing the sequences to biological or chemical carriers or coupling the sequences to tissue-type or cell-type directed ligands or antibodies
  • Compositions comprising one or more sequences and a pharmaceutically acceptable carrier are prepared by uniformly and intimately bringing into association the sequence and the pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers include liquid carriers, solid carriers or both. Liquid carriers are aqueous carriers, non-aqueous carriers or both and include, but are not limited to, aqueous suspensions, oil emulsions, water in oil emulsions, water-in-oil-in-water emulsions, site-specific emulsions, long-residence emulsions, sticky-emulsions, microemulsions and nanoemulsions. Solid carriers are biological carriers, chemical carriers or both and include, but are not limited to, viral vector systems, particles, microparticles, nanoparticles, microspheres, nanospheres, minipumps, bacterial cell wall extracts and biodegradable or non-biodegradable natural or synthetic polymers that allow for sustained release of the sequences. Methods used to complex a sequence to a solid carrier include, but are not limited to, direct adsorption to the surface of the solid carrier; covalent coupling to the surface of the solid carrier, either directly or via a linking moiety; and covalent coupling to the polymer used to make the solid carrier. Optionally, a sequence can be stabilized by the addition of non-ionic or ionic polymers such as polyoxyethylenesorbitan monooleates (Tweens) or hyaluronic acid.
  • Preferred aqueous carriers include, but are not limited to, water, saline and pharmaceutically acceptable buffers. Preferred non-aqueous carriers include, but are not limited to, a mineral oil and a neutral oil and mixtures thereof. Optionally, excipients may be included regardless of the pharmaceutically acceptable carrier used to present the sequence to the responding cells. These excipients include, but are not limited to, anti-oxidants, buffers, and bacteriostats, and may include suspending agents and thickening agents.
  • One or more sequences may be administered alone, or in combination with other therapeutic modalities including, but not limited to, chemotherapeutic agents, immunotherapeutic agents, antimicrobial agents, antiviral agents or in combination with radiation therapy. Chemotherapeutic agents include, but are not limited to, anti-metabolites, DNA damaging, microtubule destabilizing, microtubule stabilizing, actin depolymerizing, growth inhibiting, topoisomerase inhibiting, HMG-CoA inhibiting, purine inhibiting, pyrimidine inhibiting, metaloproteinase inhibiting, CDK inhibiting, angiogenesis inhibiting and differentiation enhancing agents.
  • Routes of administration include, but are not limited to, oral, topical, subcutaneous, transdermal, subdermal, intramuscular, intra-peritoneal, intra-vesical, intra-articular, intra-arterial, intra-venous, intra-dermal, intra-cranial, intra-lesional, intra-tumoral, intra-ocular, intra-pulmonary, intra-spinal, intra-prostatic, placement within cavities of the body, nasal inhalation, pulmonary inhalation, impression into skin and electrocorporation.
  • Depending on the route of administration, the volume per dose is preferably about 0.001 to 100 ml per dose, more preferably about 0.01 to 50 ml per dose and most preferably about 0.1 to 30 ml per dose. Preferably, the amount of sequence administered per dose is from about 0.001 to 100 mg/kg, more preferably from about 0.01 to 10 mg/kg and most preferably from about 0.1 to 5 mg/kg. The sequence or sequence plus a therapeutic agent can be administered in a single dose treatment, in multiple dose treatments or continuously infused on a schedule and over a period of time appropriate to the disease being treated, the condition of the recipient and the route of administration. Moreover, the sequence can be administered before, at the same time as, or after administration of the therapeutic agent.
  • A sequence in combination with chemotherapeutic agent is administered to an animal having cancer in an amount effective to potentiate the anti-neoplastic effect of the chemotherapeutic agent. Preferably, the amount of therapeutic agent administered per dose is from about 0.001 to 1000 mg/m2 or from about 0.01 to 1000 mg/kg, more preferably from about 0.01 to 500 mg/m2 or from about 0.01 to 500 mg/kg and most preferably from about 0.1 to 100 mg/m2 or from about 0.1 to 100 mg/kg. The particular sequence and the particular therapeutic agent administered, the amount per dose, the dose schedule and the route of administration should be decided by the practitioner using methods known to those skilled in the art and will depend on the type of cancer, the severity of the cancer, the location of the cancer and other clinical factors such as the size, weight and physical condition of the recipient. In addition, in vitro assays may optionally be employed to help identify optimal ranges for sequence administration and for sequence plus therapeutic agent administration.
  • Although not wanting to be bound by the following hypothesis, it is thought that the sequences of the present invention form a new family of structures and that they do not function as antisense RNAs, antisense DNAs, triple helix forming DNAs, telomerase inhibitors, transcription activators or transcription inhibitors.
  • The following examples will serve to further illustrate the present invention without, at the same time, however, constituting any limitation thereof. On the contrary, it is to be clearly understood that resort may be had to various other embodiments, modifications, and equivalents thereof which, after reading the description herein, may suggest themselves to those skilled in the art without departing from the spirit of the invention.
  • EXAMPLE 1
  • Preparation of Sequences
  • Phosphodiester and phosphorothioate sequences were prepared by HUKABEL Scientific Ltd, (Montréal, Québec, Canada) using the EXPEDITE™ 8900 automated DNA synthesis system (PersSeptive Biosystems, Inc., Farminghan, Mass.) and by Sigma-Genosys (Woodlands, Tex.) using Abacus Segmented Synthesis Technology. Unless stated otherwise, the sequences used were phosphodiester sequences. Unless stated otherwise, immediately prior to use, the sequences were dispersed in autoclaved deionized water or in an autoclaved pharmaceutically acceptable buffer such as, but not limited to, saline.
  • EXAMPLE 2
  • Cells and Reagents
  • All cell lines were obtained from the American Type Culture Collection (ATCC, Rockville, Md.) and were cultured in the medium recommended by the ATCC. Table 1 shows the cell lines, their origins and their properties.
    TABLE 1
    Cell lines
    CELL LINE ORIGIN PROPERTIES
    THP-1 Human acute monocytic leukemia Suspension tumor model
    p53 null
    MCF-7 Human breast cancer Solid tumor model; non-metastatic
    Caspase 3-negative; estrogen-dependent
    JURKAT Human T cell leukemia Suspension tumor model
    Atypical multi-drug resistance
    associated with p190-MRP protein
    PC-3 Human prostate cancer Solid tumor model; metastatic
    p53 mutated; androgen-independent
    (hormone refractory)
    LNCaP Human prostate cancer Solid tumor model; metastatic
    TGF-beta 1 receptor-negative;
    androgen-dependent
    OVCAR-3 Human ovarian cancer Solid tumor model; metastatic
    p53 mutated; p21/waf-1/Cip-1 deleted
    SK-OV-3 Human ovarian cancer Solid tumor model; metastatic
    p53 deleted; p21/waf-1/Cip deleted;
    p15ink4B, p16ink4 deleted
    HL-60 Human promyelocytic leukemia Suspension tumor model
    p53 Mutated
    EL-4 Murine T lymphoma Suspension tumor model
    A20 Murine B cell leukemia Suspension tumor model
    L-1210 Murine leukemia Suspension tumor model
    D-17 Canine osteosarcoma Solid tumor model
    CF-51 Canine mammary gland cancer Solid tumor model
  • Cells were seeded in 6 (1 ml/well), 24 (0.5 ml/well) or 96 (0.1 ml/well) well flat-bottom microplates and were maintained at 37° C. in a 5% CO2 atmosphere. Unless stated otherwise, 2.5×105 cells/ml were incubated with 0 μg/ml (control) and 100 μg/ml (treated) of 2 to 45 base sequences containing A, C, G and T for 48 h.
  • EXAMPLE 3
  • Measurement of Cell Proliferation
  • Cell proliferation was measured using dimethylthiazol-diphenyl-tetrazolium (MTT) reduction (Mosman et al. J. Immunol. Methods 65:55, 1983). MTT was measured at a wavelength of 570 nm using a multiplate spectrophotometer reader (ELX800, Bio-TEK Instruments Inc., Winooski, Vt.).
  • EXAMPLE 4
  • Inhibition of Jurkat Human Leukemia T Cell Proliferation
  • Jurkat human leukemia T cells are an atypical multi-drug resistant human suspension tumor cell model. Jurkat cells were incubated with 27 base GT and CT sequences (Table 2).
    TABLE 2
    % inhibition of Jurkat human leukemia
    T cell proliferation
    SEQUENCE % INHIBITION
    GTGTGTGTGTGTGTGTGTGTGTGTGTG-(GT)13G 51
    SEQ ID NO:1-(27 bases)
    GGGTGGGTGGGTGGGTGGGTGGGTGGG-(G3T)6G3 23
    SEQ ID NO:2-(27 bases)
    GGGGGTGGGGGTGGGGGTGGGGGTGGG-(G5T)4G3 24
    SEQ ID NO:3-(27 bases)
    GGGGGGGTGGGGGGGTGGGGGGGTGG-(G7T)3G3 11
    SEQ ID NO:4-(27 bases)
    TGTGTGTGTGTGTGTGTGTGTGTGTG-(TG)13T 45
    SEQ ID NO:5-(27 bases)
    TCTCTCTCTCTCTCTCTCTCTCTCTCT-(TC)13 T 0
    SEQ ID NO:6-(27 bases)

    As shown in Table 2, Jurkat T cell proliferation was inhibited by the GT sequences tested, but not by the CT sequence tested.
  • Jurkat T cells were incubated with 3, 6, 9, 12, 14, 15, 18, 21 and 24 base GT sequences (Table 3).
    TABLE 3
    % inhibition of Jurkat human leukemia
    T cell proliferation
    SEQUENCE % INHIBITION
    TGT-(TG)1T 22
    SEQ ID NO:7-(3 bases)
    GTG-(GT)1G 46
    SEQ ID NO:8-(3 bases)
    TGTGTG-(TG)3 36
    SEQ ID NO:9-(6 bases)
    GTGTGT-(GT)3 48
    SEQ ID NO:10-(6 bases)
    TGTGTGTGT-(TG)4T 45
    SEQ ID NO:11-(9 bases)
    GTGTGTGTG-(GT)4G 47
    SEQ ID NO:12-(9 bases)
    TGTGTGTGTGTG-(TG)6 49
    SEQ ID NO:13-(12 bases)
    GTGTGTGTGTGT-(GT)6 51
    SEQ ID NO:14-(12 bases)
    TGTGTGTGTGTGTG-(TG)7 47
    SEQ ID NO:15-(14 bases)
    GTGTGTGTGTGTGTG-(GT)7G 58
    SEQ ID NO:16-(15 bases)
    TGTGTGTGTGTGTGTGTG-(TG)9 56
    SEQ ID NO:17-(18 bases)
    GTGTGTGTGTGTGTGTGT-(GT)9 60
    SEQ ID NO:18-(18 bases)
    TGTGTGTGTGTGTGTGTGTGT-(TG)10T 60
    SEQ ID NO:19-(21 bases)
    GTGTGTGTGTGTGTGTGTGTG-(GT)10G 46
    SEQ ID NO:20-(21 bases)
    TGTGTGTGTGTGTGTGTGTGTGTG-(TG)12 54
    SEQ ID NO:21-(24 bases)
    GTGTGTGTGTGTGTGTGTGTGTGT-(GT)12 56
    SEQ ID NO:22-(24 bases)

    As shown in Table 3, 3, 6, 9, 12, 14, 15 and 18 base GT sequences inhibited Jurkat T cell proliferation as effectively as 21 and 24 bases GT sequences.
  • Jurkat T cells were incubated with 6 base GT, AG, CG, GG, AGT and CGT sequences (Table 4).
    TABLE 4
    % inhibition Jurkat human leukemia
    T cell proliferation
    SEQUENCE % INHIBITION
    TGTGTG-(TG)3 36
    SEQ ID NO:9-(6 bases)
    GTGTGT-(GT)3 48
    SEQ ID NO:10-(6 bases)
    TTTGTT-TT(TG)1TT 31
    SEQ ID NO:23-(6 bases)
    GGTGGG-GG(TG)1GG 48
    SEQ ID NO:24-(6 bases)
    GGGTGG-GG(GT)1GG 60
    SEQ ID NO:25-(6 bases)
    TTGTTT-TT(GT)1TT 34
    SEQ ID NO:26-(6 bases)
    AAGTAA-AA(GT)1AA 13
    SEQ ID NO:27-(6 bases)
    CCGTCC-CC(GT)1CC 11
    SEQ ID NO:28-(6 bases)
    TGGTTG-TG(GT)1TG 42
    SEQ ID NO:29-(6 bases)
    ATGTAT-AT(GT)1AT 16
    SEQ ID NO:30-(6 bases)
    AGGTGA-AG(GT)1GA 10
    SEQ ID NO:31-(6 bases)
    GAGTGA-GA(GT)1GA 24
    SEQ ID NO:32-(6 bases)
    GGGTCT-GG(GT)1CT 15
    SEQ ID NO:33-(6 bases)
    CCGTGG-CC(GT)1GG 37
    SEQ ID NO:34-(6 bases)
    GGGTCC-GG(GT)1CC 20
    SEQ ID NO:35-(6 bases)
    CTGTCT-CT(GT)1CT 19
    SEQ ID NO:36-(6 bases)
    TCGTTC-TC(GT)1TC 20
    SEQ ID NO:37-(6 bases)
    CGGTGC-CG(GT)1GC 16
    SEQ ID NO:38-(6 bases)
    TTGTGG-TT(GT)1GG 35
    SEQ ID NO:39-(6 bases)
    GGGTTT-GG(GT)1TT 31
    SEQ ID NO:40-(6 bases)
    GGTTGG-GG(TT)1GG 43
    SEQ ID NO:41-(6 bases)
    GGAAGG-GG(AA)1GG 22
    SEQ ID NO:42-(6 bases)
    GGCCGG-GG(CC)GG 29
    SEQ ID NO:43-(6 bases)
    GGGGGG-GG(GG)1GG 26
    SEQ ID NO:44-(6 bases)
    GGGAGG-GG(GA)1GG 28
    SEQ ID NO:45-(6 bases)
    GGGCGG-GG(GC)1GG 23
    SEQ ID NO:46-(6 bases)
    GGAGGG-GG(AG)1GG 14
    SEQ ID NO:47-(6 bases)
    GTGGGG-(GT)1G4 26
    SEQ ID NO:48-(6 bases)
    TTAGGG-TT(AG)1GG 45
    SEQ ID NO:49-(6 bases)
  • As shown in Table 4, 6 base GT sequences inhibited Jurkat T cell proliferation. GT SEQ ID NO:25 inhibited proliferation 60% and AGT SEQ ID NO:49 inhibited proliferation 45%. Comparison of the relative potency of GT SEQ ID NO:25 and AGT SEQ ID NO:49 using PHARM/PCS-4 Software (Microcomputer Specialists, Philadelphia, Pa.) showed the potency of GT SEQ ID NO:25 to be 3.4 times that of AGT SEQ ID NO:49. AGT SEQ ID NO:49-phosphorothioate is reported to inhibit telomerase activity and to induce apoptosis in Burkitt lymphoma cells (Mata et al. Toxicol. Appl. Pharmacol. 144:189, 1997).
  • To determine telomerase activity, extracts from 2×105 Jurkat T cells were assayed using the PCR-telomeric repeat amplification protocol (TRAP) (Roche, Laval, Québec, Canada). At concentrations between 1 and 100 μg/ml, GT SEQ ID NO:25-phosphodiester showed between 0 and 10% anti-telomerase activity, whereas AGT SEQ ID NO:49-phosphorothioate showed between 30 and 75% anti-telomerase activity. Neither GT SEQ ID NO:25-phosphorothioate nor GT SEQ ID NO:49-phosphodiester showed any anti-telomerase actvity.
  • Jurkat T cells were incubated with 2, 3, 4, 5, 6 and 7 base GT sequences (Table 5).
    TABLE 5
    % inhibition of Jurkat human leukemia
    T cell proliferation
    SEQUENCE % INHIBITION
    GT-(GT)1 38
    SEQ ID NO:50-(2 bases)
    TG-(TG)1 50
    SEQ ID NO:51-(2 bases)
    TGT-(TG)1T 22
    SEQ ID NO:7 (3 bases)
    GTG-(GT)1G 46
    SEQ ID NO:8-(3 bases)
    GTGG-G(TG)1G 52
    SEQ ID NO:52-(4 bases)
    TTGT-T(GT)1T 25
    SEQ ID NO:53-(4 bases)
    GTGT-G(TG)1T 42
    SEQ ID NO:54-(4 bases)
    TTGG-T(TG)1G 44
    SEQ ID NO:55-(4 bases)
    GGTG-G(GT)1G 54
    SEQ ID NO:56-(4 bases)
    TGTT-T(GT)1T 32
    SEQ ID NO:57-(4 bases)
    GGTT-G(GT)1T 37
    SEQ ID NO:58-(4 bases)
    TGTG-T(GT)1G 52
    SEQ ID NO:59-(4 bases)
    GGTGG-G(GT)1G2 50
    SEQ ID NO:60-(5 bases)
    GGGTG-G2(GT)G 50
    SEQ ID NO:61-(5 bases)
    TGTGTG-(TG)3 36
    SEQ ID NO:9-(6 bases)
    GTGTGT-(GT)3 48
    SEQ ID NO:10-(6 bases)
    TTTGTT-TT(TG)1TT 31
    SEQ ID NO:23-(6 bases)
    GGTGGG-GG(TG)1GG 48
    SEQ ID NO:24-(6 bases)
    GGGTGG-GG(GT)1GG 60
    SEQ ID NO:25-(6 bases)
    TTGTTT-TT(GT)1TT 34
    SEQ ID NO:26-(6 bases)
    TGGTTG-TG(GT)1TG 42
    SEQ ID NO:29-(6 bases)
    GGGGTGG-G3(GT)1G2 41
    SEQ ID NO:62-(7 bases)
    GGGTGGG-G2(GT)G3 28
    SEQ ID NO:63-(7 bases)
    TGGGTGG-TG2(GT)1G2 55
    SEQ ID NO:64-(7 bases)
    GGGTGGT-G2(GT)1G2T 48
    SEQ ID NO:65-(7 bases)

    As shown in Table 5, 2, 3, 4, 5, 6 and 7 base GT sequences inhibited Jurkat T cell proliferation.
  • EXAMPLE 5
  • Inhibition of HL-60 Human Promyelocytic Leukemia Cell Proliferation
  • HL-60 promyelocytic leukemia cells are a p53 mutated human suspension tumor model. HL-60 cells were incubated with 6 base GT sequences (Table 6).
    TABLE 6
    % inhibition of HL-60 human promyelocytic
    leukemia cell proliferation
    SEQUENCE % INHIBITION
    TGTGTG-(TG)3 7
    SEQ ID NO:9-(6 bases)
    GTGTGT-(GT)3 13
    SEQ ID NO:10-(6 bases)
    TTTGTT-TT(TG)1TT 13
    SEQ ID NO:23-(6 bases)
    GGTGGG-GG(TG)1GG 18
    SEQ ID NO:24-(6 bases)
    GGGTGG-GG(GT)1GG 35
    SEQ ID NO:25-(6 bases)
    TTGTTT-TT(GT)1TT 16
    SEQ ID NO:26-(6 bases)

    As shown in Table 6, 6 base GT sequences inhibited HL-60 cell proliferation.
  • EXAMPLE 6
  • Inhibition of MCF-7 Human Breast Cancer Cell Proliferation
  • MCF-7 human breast cancer cells are a caspase 3 negative, estrogen-dependent human solid tumor model. MCF-7 cells (5×105 cells/ml) were incubated with 3 and 6 base GT sequences (Table 7).
    TABLE 7
    % inhibition of MCF-7 human breast
    cancer cell proliferation
    SEQUENCE % INHIBITION
    TGT-(TG)T −6
    SEQ ID NO:7-(3 bases)
    GTG-(GT)G 18
    SEQ ID NO:8-(3 bases)
    TGTGTG-(TG)3 6
    SEQ ID NO:9-(6 bases)
    GTGTGT-(GT)3 31
    SEQ ID NO:10-(6 bases)
    TTTGTT-TT(TG)1 TT 7
    SEQ ID NO:23-(6 bases)
    GGTGGG-GG(TG)1GG 41
    SEQ ID NO:24-(6 bases)
    GGGTGG-GG(GT)1GG 41
    SEQ ID NO:25-(6 bases)
    TTGTTT-TT(GT)1 TT 20
    SEQ ID NO:26-(6 bases)

    As shown in Table 7, 6 and 7 base GT sequences inhibited MCF-7 cell proliferation.
  • EXAMPLE 7
  • Inhibition of PC-3 Human Prostate Cancer Cell Proliferation
  • PC-3 prostate cancer cells are a p53 mutated, androgen-independent human solid tumor model. PC-3 cells (5×105 cells/ml) were incubated with 3 and 6 base GT sequences (Table 8).
    TABLE 8
    % inhibition of PC-3 human prostate
    cancer cell proliferation
    SEQUENCE % INHIBITION
    TGT-(TG)T 8
    SEQ ID NO:7-(3 bases)
    GTG-(GT)G 13
    SEQ ID NO:8-(3 bases)
    TGTGTG(TG)3 16
    SEQ ID NO:9-(6 bases)
    GTGTGT-(GT)3 37
    SEQ ID NO:10-(6 bases)
    TTTGTT-TT(TG)1TT 14
    SEQ ID NO:23-(6 bases)
    GGTGGG-GG(TG)1GG 26
    SEQ ID NO:24-(6 bases)
    GGGTGG-GG(GT)1GG 38
    SEQ ID NO:25-(6 bases)
    TTGTTT-TT(GT)1TT 18
    SEQ ID NO:26-(6 bases)

    As shown in Table 8, 3 and 6 base GT sequences inhibited PC-3 cell proliferation.
  • EXAMPLE 8
  • Inhibition of LNCaP Human Prostate Cancer Cell Proliferation
  • LNCaP prostate cancer cells are a TGF-beta 1 receptor negative, androgen-independent, metastatic human solid tumor model. LNCaP cells (5×105 cells/ml) were incubated with 6 base GT sequences (Table 9).
    TABLE 9
    % inhibition of LNCaP human prostate
    cancer cell proliferation
    SEQUENCE % INHIBITION
    TGTGTG-(TG)3 −9
    SEQ ID NO:9-(6 bases)
    GTGTGT-(GT)3 −4
    SEQ ID NO:10-(6 bases)
    TTTGTT-TT(TG)TT 14
    SEQ ID NO:23-(6 bases)
    GGTGGG-GG(TG)GG 17
    SEQ ID NO:24-(6 bases)
    GGGTGG-GG(GT)GG 18
    SEQ ID NO:25-(6 bases)
    TTGTTT-TT(GT)TT 22
    SEQ ID NO:26-(6 bases)

    As shown in Table 9, 6 base GT sequences inhibited LNCaP cell proliferation.
  • EXAMPLE 9
  • Inhibition of THP-1 Human Acute Monocytic Leukemia Cell Proliferation
  • THP-1 acute monocytic leukemia cells are a p53 null human suspension tumor model. THP-1 cells (1.6×106 cells/ml) were incubated with 6 base GT sequences (Table 10).
    TABLE 10
    % inhibition of THP-1 human acute
    monocytic leukemia cell proliferation
    SEQUENCE % INHIBITION
    TGTGTG-(TG)3 0
    SEQ ID NO:9-(6 bases)
    GTGTGT-(GT)3 11
    SEQ ID NO:10-(6 bases)
    TTTGTT-TT(TG)1TT 8
    SEQ ID NO:23-(6 bases)
    GGTGGG-GG(TG)1GG 6
    SEQ ID NO:24-(6 bases)
    GGGTGG-GG(GT)1GG 15
    SEQ ID NO:25-(6 bases)
    TTGTTT-TT(GT)1 TT 1
    SEQ ID NO:26-(6 bases)

    As shown in Table 10, 6 base GT sequences inhibited THP-1 cell proliferation.
  • EXAMPLE 10
  • Inhibition of OVCAR-3 Human Ovarian Cancer Cell Proliferation
  • OVCAR-3 ovarian cancer cells are a p53 mutated, p21/waf-1/Cip deleted, metastatic human solid tumor model. OVCAR-3 cells (5×105 cells/ml) were incubated with 2, 6 and 18 base GT sequences and with a 6 base AGT sequence (Table 11).
    TABLE 11
    % inhibition of OVCAR-3 human
    ovarian cancer cell proliferation
    SEQUENCE % INHIBITION
    TG-(TG)1 23
    SEQ ID NO:51-(2 bases)
    TTAGGG-TT(AG)1GG 15
    SEQ ID NO:49-(6 bases)
    GTGTGTGTGTGTGTGTGT-(GT)9 10
    SEQ ID NO:18-(18 bases)
    GGGTGG-GG(GT)1GG 15
    SEQ ID NO:25-(6 bases)

    As shown in Table 11, 2, 6 and 18 base GT sequences and a 6 base AGT sequence inhibited OVCAR-3 cell proliferation.
  • EXAMPLE 11
  • Inhibition of SK-OV-3 Human Ovarian Cancer Cell Proliferation
  • SK-OV-3 ovarian cancer cells are a p53 mutated, p21/waf-1/Cip deleted, p15ink4B deleted, p16Ink4 deleted, metastatichuman solid tumor model. SK-OV-3 cells (5×105 cells/ml) were incubated with 2, 6 and 18 base GT sequences (Table 12).
    TABLE 12
    % inhibition of SK-OV-3 human ovarian cancer
    cell proliferation
    SEQUENCE % INHIBITION
    TG-(TG)1 18
    SEQ ID NO:51-(2 bases)
    TTAGGG-TT(AG)1GG 11
    SEQ ID NO:49-(6 bases)
    GTGTGTGTGTGTGTGTGT-(GT)9 6
    SEQ ID NO:18-(18 bases)
    GGGTGG-GG(GT)1GG 12
    SEQ ID NO:25-(6 bases)

    As shown in Table 12, 2, 6 and 18 base GT sequences inhibited SK-OV-3 cell proliferation.
  • EXAMPLE 12
  • Inhibition of Cell Proliferation by Phosphodiester and Phosphorothioate Sequences
  • Modification of natural phosphodiester sequences by substitution of a sulfur atom for a nonbridging oxygen atom on one or more of the phosphate groups has been reported to increase the stability of oligonucleotide sequences to endonucleases in biological fluids and cells (Crooke et al. Anticancer Drugs 6:609, 1991).
  • Jurkat human leukemia T cells (Table 13), LNCaP human prostate cancer cells (5×105 cells/ml) (Table 14) and MCF-7 human breast cancer cells (5×105 cells/ml) (Table 15) were incubated with 6 base GT sequences, having either oxygen (phosphodiester) or sulfur (phosphorothioate) as a nonbridging atom on the phosphate group.
    TABLE 13
    % inhibition of Jurkat human leukemia T cell
    proliferation
    % INHIBITION
    SEQUENCE PHOSPHODIESTER PHOSPHOROTHIOATE
    TGTGTG-(TG)3 37 −17
    (6 bases)
    SEQ ID NO:9
    phosphodiester;
    phosphorothioate
    GTGTGT-(GT)3 44 0
    (6 bases)
    SEQ ID NO:10
    phosphodiester;
    phosphorothioate
    TTTGTT-TT(TG)1TT 31 4
    (6 bases)
    SEQ ID NO:23
    phosphodiester;
    phosphorothioate
    GGTGGG-GG(TG)1GG 48 6
    (6 bases)
    SEQ ID NO:24
    phosphodiester;
    phosphorothioate
    GGGTGG-GG(GT)1GG 60 0
    (6 bases)
    SEQ ID NO:25
    phosphodiester;
    phosphorothioate
    TTGTTT-TT(GT)1TT 34 0
    (6 bases)
    SEQ ID NO:26
    phosphodiester;
    phosphorothioate
  • TABLE 14
    % inhibition of LNCaP human prostate cancer
    cell proliferation
    % INHIBITION
    SEQUENCE PHOSPHODIESTER PHOSPHOROTHIOATE
    TGTGTG-(TG)3 −9 −16
    (6 bases)
    SEQ ID NO:9
    phosphodiester;
    phosphorothioate
    GTGTGT-(GT)3 −4 −20
    (6 bases)
    SEQ ID NO:10
    phosphodiester;
    phosphorothioate
    TTTGTT-TT(TG)1TT 14 −11
    (6 bases)
    SEQ ID NO:23
    phosphodiester;
    phosphorothioate
    GGTGGG-GG(TG)1GG 17 −17
    (6 bases)
    SEQ ID NO:24
    phosphodiester;
    phosphorothioate
    GGGTGG-GG(GT)1GG 18 −8
    (6 bases)
    SEQ ID NO:25
    phosphodiester;
    phosphorothioate
    TTGTTT-TT(GT)1TT 22 −1
    (6 bases)
    SEQ ID NO:26
    phosphodiester;
    phosphorothioate
  • TABLE 15
    % inhibition of MCF-7 human breast cancer
    cell proliferation
    % INHIBITION
    SEQUENCE PHOSPHODIESTER PHOSPHOROTHIOATE
    TGTGTG-(TG)3 6 6
    (6 bases)
    SEQ ID NO:9
    phosphodiester;
    phosphorothioate
    GTGTGT-(GT)3 31 12
    (6 bases)
    SEQ ID NO:10
    phosphodiester;
    phosphorothioate
    TTTGTT-TT(TG)1 TT 7 8
    (6 bases)
    SEQ ID NO:23
    phosphodiester;
    phosphorothioate
    GGTGGG-GG(TG)1GG 41 12
    (6 bases)
    SEQ ID NO:24
    phosphodiester;
    phosphorothioate
    GGGTGG-GG(GT)1GG 41 12
    (6 bases)
    SEQ ID NO:25
    phosphodiester;
    phosphorothioate
    TTGTTT-TT(GT)1 TT 20 6
    (6 bases)
    SEQ ID NO:26
    phosphodiester;
    phosphorothioate

    As shown in Tables 13, 14 and 15, 6 base GT-phosphodiester sequences inhibited Jurkat T, LNCaP and MCF-7 cell proliferation more effectively than 6 base GT-phosphorothioate sequences.
  • EXAMPLE 13
  • Inhibition of Cell Proliferation by Mixed Phosphodiester and Phosphorothioate Sequences
  • Jurkat human leukemia T cells (Table 16) and MCF-7 human breast cancer cells (5×105 cells/ml) (Table 17) were incubated with the 6 base GT SEQ ID NO:25, wherein either oxygen (phosphodiester) or sulfur (phosphorothioate) was the nonbridging atom on the phosphate group.
    TABLE 16
    % inhibition of Jurkat human leukemia T cell
    proliferation
    %
    SEQUENCE* INHIBITION
    GoGoGoToGoGo-GoGo(GoTo)1GoGo (oxygen 60
    atom 1 to 6)
    SEQ ID NO:25-(6 bases)
    GoGoGsToGoGo-GoGo(GsTo)1GoGo (oxygen 17
    atom 1, 2, 4, 5, 6; sulfur atom 3)
    SEQ ID NO:25-(6 bases)
    GoGoGoTsGoGo-GoGo(GoTs)1GoGo (oxygen 12
    atom 1, 2, 3, 5, 6; sulfur atom 4)
    SEQ ID NO:25-(6 bases)
    GoGoGsTsGoGo-GoGo(GsTs)1GoGo (oxygen 13
    atom 1, 2, 5, 6; sulfur atom 3, 4)
    SEQ ID NO:25-(6 bases)
    GsGoGoToGoGs-GsGo(GoTo)1GoGs (oxygen 16
    atom 2, 3, 4, 5; sulfur atom 1, 6)
    SEQ ID NO:25-(6 bases)
    GoGsGoToGsGo-GoGs(GoTo)1GsGo (oxygen 11
    atom 1, 3, 4, 6; sulfur atom 2, 5)
    SEQ ID NO:25-(6 bases)
    GsGsGoToGsGs-GsGs(GoTo)1GsGs (oxygen −13
    atom 3, 4; sulfur atom 1, 2, 5, 6)
    SEQ ID NO:25-(6 bases)
    GsGsGsTsGsGs-GsGs(GsTs)1GsGs (sulfur 0
    atom 1 to 6)
    SEQ ID NO:25-(6 bases;
    phosphorothioate)

    *Note:

    o” represents an oxygen atom and “s” represents a sulfur atom on the phosphate group
  • TABLE 17
    % inhibition of MCF-7 human breast cancer cell
    proliferation
    %
    SEQUENCE* INHIBITION
    GoGoGoToGoGo-GoGo(GoTo)1GoGo (oxygen 41
    atom 1 to 6)
    SEQ ID NO:25-(6 bases;
    phosphodiester)
    GoGoGsToGoGo-GoGo(GsTo)1GoGo (oxygen 12
    atom 1, 2, 4, 5, 6; sulfur atom 3)
    SEQ ID NO:25-(6 bases)
    GoGoGoTsGoGo-GoGo(GoTs)1GoGo (oxygen 0
    atom 1, 2, 3, 5, 6; sulfur atom 4)
    SEQ ID NO:25-(6 bases)
    GoGoGsTsGoGo-GoGo(GsTs)1GoGo (oxygen 43
    atom 1, 2, 5, 6; sulfur atom 3, 4)
    SEQ ID NO:25-(6 bases)
    GsGoGoToGoGs-GsGo(GoTo)1GoGs (oxygen 12
    atom 2, 3, 4, 5; sulfur atom 1, 6)
    SEQ ID NO:25-(6 bases)
    GoGsGoToGsGo-GoGs(GoTo)1GsGo (oxygen 13
    atom 1, 3, 4, 6; sulfur atom 2, 5)
    SEQ ID NO:25-(6 bases)
    GsGsGoToGsGs-GsGs(GoTo)1GsGs (oxygen −3
    atom 3, 4; sulfur atom 1, 2, 5, 6)
    SEQ ID NO:25-(6 bases)
    GsGsGsTsGsGs-GsGs(GsTs)1GsGs (sulfur 12
    atom 1 to 6)
    SEQ ID NO:25-(6 bases;
    phosphorothioate)

    *Note:

    o” represents an oxygen atom and “s” represents a sulfur atom on the phosphate group

    As shown in Tables 16 and 17, substitution of a sulfur atom for a nonbridging oxygen atom on one or more phosphate groups of 6 base GT SEQ ID NO:25 resulted in a significant decrease in inhibition of Jurkat T and MCF-7 cell proliferation.
  • EXAMPLE 14
  • Inhibition of Murine Cancer Cell Proliferation
  • EL-4 murine lymphoma T cells are a suspension tumor model. EL-4 murine lymphoma T cells were incubated with 6, 18, 27 and 33 base GT sequences and with a 15 base ACG sequence (Table 18).
    TABLE 18
    % inhibition of EL-4 murine T lymphoma cell
    proliferation
    SEQUENCE % INHIBITION
    GGGTGG-GG(GT)1GG 4
    SEQ ID NO:25-(6 bases)
    GGGTGG-GG(GT)1GG −8
    SEQ ID NO:25-(6 bases
    phosphorothioate)
    GTGTGTGTGTGTGTGTGTGTGTGTGTG-(G1T)13 G 1
    SEQ ID NO:1-(27 bases)
    GTGTGTTTGGTGGTTTTGTTTGTTGTTTTTTTG −1
    SEQ ID NO:66-(33 bases)
    AACCACAAGCCCAAC −6
    SEQ ID NO:67-(15 bases)
    GTGTGT-(GT)3 −2
    SEQ ID NO:10-(6 bases)
    GTGTGTGTGTGTGTGTGT-(GT)9 −2
    SEQ ID NO:18-(18 bases)

    As shown in Table 18, 6, 18, 27 and 33 base GT sequences and a 15 base ACG sequence did not inhibit EL-4 murine cell proliferation.
  • A20 murine leukemia B cells are a suspension tumor model. A20 murine leukemia B cells were incubated with 6 base GT sequences (Table 19).
    TABLE 19
    % inhibition of A20 murine B leukemia cell
    proliferation
    SEQUENCE % INHIBITION
    TGTGTG-(TG)3 22
    SEQ ID NO:9-(6 bases)
    GTGTGT-(GT)3 9
    SEQ ID NO:10-(6 bases)
    TTTGTT-TT(TG)1TT 5
    SEQ ID NO:23-(6 bases)
    GGTGGG-GG(TG)1GG 9
    SEQ ID NO:24-(6 bases)
    GGGTGG-GG(GT)1GG 11
    SEQ ID NO:25-(6 bases)
    TTGTTT-TT(GT)1TT 15
    SEQ ID NO:26-(6 bases)

    As shown in Table 19, 6 base GT sequences inhibited proliferation of A20 murine B leukemia cells.
  • EXAMPLE 15
  • Inhibition of Canine Cancer Cell Proliferation
  • D-17 canine osteosarcoma cells and CF-51 canine mammary gland cancer cells are solid tumor models. D-17 canine osteosarcoma cells (5×105 cells/ml) (Table 20) and CF-51 canine mammary gland cancer cells (5×105 cells/ml) (Table 21) were incubated with 6, 9, 17, 27 and 33 base GT-sequences and with a 15 base ACG sequence.
    TABLE 20
    % inhibition of D-17 canine osteosarcoma
    cell proliferation
    SEQUENCE % INHIBITION
    GGGTGG-GG(GT)1GG 18
    SEQ ID NO:25-(6 bases)
    GTGTGTGTGTGTGTGTGTGTGTGTGTG-(GT)13G 23
    SEQ ID NO:1-(27 bases)
    GTGTGTTTGGTGGTTTTGTTTGTTGTTTTTTTG 23
    SEQ ID NO:66-(33 bases)
    AACCACAAGCCCAAC 20
    SEQ ID NO:67-(15 bases)
    GTGTGT-(GT)3 15
    SEQ ID NO:10-(9 bases)
    TGTGTGTGTGTGTGTGT-(TG)8T 8
    SEQ ID NO:17-(17 bases)
  • TABLE 21
    % inhibition of CF-51 canine mammary gland
    cancer cell proliferation
    SEQUENCE % INHIBITION
    GGGTGG-GG(GT)1GG 14
    SEQ ID NO:25-(6 bases)
    GTGTGTGTGTGTGTGTGTGTGTGTGTG-(GT)13G 23
    SEQ ID NO:1-(27 bases)
    GTGTGTTTGGTGGTTTTGTTTGTTGTTTTTTTG 23
    SEQ ID NO:66-(33 bases)
    AACCACAAGCCCAAC 20
    SEQ ID NO:67-(15 bases)
    GTGTGT-(GT)3 15
    SEQ ID NO:10-(9 bases)
    TGTGTGTGTGTGTGTGT-(TG)8T 8
    SEQ ID NO:17-(17 bases)

    As shown in Tables 20 and 21, 6, 9, 17, 27 and 33 base GT sequences and a 15 base ACG sequence inhibited both D-17 and CF-51 canine cell proliferation.
  • EXAMPLE 16
  • Inhibition of Cancer Cell Proliferation
  • Inhibition of human, murine and canine cancer cell proliferation by 6 base GT SEQ ID NO:25 is summarized in Table 22.
    TABLE 22
    % inhibition of human, murine and canine cancer cell proliferation
    CELLS GG(GT)1GG (SEQ ID NO: 25)
    HUMAN 60
    Jurkat
    HUMAN 38
    PC-3
    HUMAN 41
    MCF-7
    HUMAN 35
    HL-60
    HUMAN 14
    OVCAR-3
    HUMAN 18
    LNCaP
    HUMAN 12
    SK-OV-3
    HUMAN 15
    THP-1
    MURINE 1
    EL-4
    MURINE 11
    A20
    MURINE 8
    L-1210
    CANINE 18
    D17
    CANINE 14
    CF-51

    As shown in Table 22, human cancer cells are more sensitive than canine cancer cells and murine cancer cells to inhibition of proliferation by 6 base GT SEQ ID NO:25.
  • EXAMPLE 17
  • Synergistic Effect of Two 6 Base GTSequences on Inhibition of Proliferation
  • Jurkat human leukemia T cells were incubated with suboptimal concentrations (5.0 μg/ml) of 6 base GT sequences (Table 23).
    TABLE 23
    % inhibition of Jurkat human leukemia
    T cell proliferation
    SEQUENCE % INHIBITION
    GGGTGG-GG(GT)1GG 5
    SEQ ID NO:25-(6 bases)
    TTGTTT-TT(GT)1GG −2
    SEQ ID NO:26-(6 bases)
    GG(GT)1GG + TT(GT)1GG 14
    SEQ ID NO:25 + SEQ ID NO:26
    TGGTTG-TG(GT)1TG −1
    SEQ ID NO:29-(6 bases)
    TGGTTG-TG(GT)1 TG 2
    SEQ ID NO:10-(6 bases)
    TG(GT)1TG + TG(GT)1TG 9
    SEQ ID NO:29 + SEQ ID NO:10
    GGTTGG-GG(TT)1GG 4
    SEQ ID NO:41-(6 bases)
    TTGTGG-TT(GT)1GG 4
    SEQ ID NO:39-(6 bases)
    GG(TT)1GG + TT(GT)1GG 18
    SEQ ID NO:41 + SEQ ID NO:39

    As shown in Table 23, the simultaneous use of two 6 base GT sequences had a synergistic effect on inhibition of Jurkat T cell proliferation.
  • EXAMPLE 18
  • Potentiation of Antineoplastic Effect of Chemotherapeutic Drugs
  • Jurkat human leukemia T cells were incubated with 1.0 μg/ml of 6 base GT SEQ ID NO:25 and of 27 base GT SEQ ID NO:1 in the presence of 0, 0.1, 1.0 or 10.0 μg/ml of 5-fluorouracil or cisplatin (Table 24). 5-fluorouracil is an antimetabolite that interferes with DNA and RNA synthesis. Cisplatin. is an alkylating agent that cross-links DNA and inhibits DNA precursors.
    TABLE 24
    % inhibition of Jurkat human leukemia T cell proliferation
    % INHIBITION
    SEQUENCES 0.0 0.1 1.0 10.0
    5-Fluorouracil (μg/ml)
    5-Fluorouracil 0 3 14 38
    GG(GT)1GG-(6 bases) 0 10 21 40
    SEQ ID NO: 25 at 1.0 μg/ml
    (GT)13G-(27 bases) 3 15 25 41
    SEQ ID NO: 1 at 1.0 μg/ml
    Cisplatin (μg/ml)
    Cisplatin 0 7 29 73
    GG(GT)1GG-(6 bases) 0 14 38 76
    SEQ ID NO: 25 at 1.0 μg/ml
    (GT)13G-(27 bases) 3 18 35 76
    SEQ ID NO: 1 at 1.0 μg/ml
  • As shown in Table 24, 6 base GT SEQ ID NO:25 and 27 base GT SEQ ID NO:1 potentiated the antineoplastic effect of 0.1 and 1.0 μg/ml of 5-fluorouracil on Jurkat T cell proliferation and GT SEQ ID NO:25 potentiated the antineoplastic effect of 0.1 and 1.0 μg/ml cisplatin on Jurkat T cell proliferation.
  • MCF-7 human breast cancer cells (5×105 cells/ml) were incubated with 1.0 μg/ml of 6 base GT SEQ ID NO:25 and of 27 base GT SEQ ID NO:1 in the presence of 0, 0.1, 1.0 or 10.0 μg/ml of 5-fluorouracil or tamoxifen (Table 25). Tamoxifen is an estrogen antagonist.
    TABLE 25
    % inhibition of MCF-7 human breast cancer cell proliferation
    % INHIBITION
    SEQUENCES 0.0 0.1 1.0 10.0
    5-luorouracil (μg/ml)
    5-Fluorouracil 0 13 28 28
    GG(GT)1GG-(6 bases) 6 24 36 33
    SEQ ID NO: 25 at 1.0 μg/ml
    (G1T)13G (27 bases) 8 24 35 33
    SEQ ID NO: 1 at 1.0 μg/ml
    Tamoxifen (μg/ml)
    Tamoxifen 0 10 18 15
    GG(GT)1GG-(6 bases) 6 21 24 31
    SEQ ID NO: 25 at 1.0 μg/ml
    (GT)13G-(27 bases) 8 19 24 20
    SEQ ID NO: 1 at 1.0 μg/ml
  • As shown in Table 25, 6 base SEQ ID NO:25 potentiated the antineoplastic effect of 0.1 μg/ml 5-flurouracil and of 0.1 μg/ml tamoxifen on MCF-7 cell proliferation. Twenty-seven base SEQ ID NO:1 did not potentiate the antineoplastic activity of 5-fluorouracil or of tamoxifen on MCF-7 cell proliferation.
  • EXAMPLE 19
  • Inhibition of Proliferation by Repeats of 6 Base GT SEQ ID NO:25
  • Jurkat human leukemia T cells were incubated with 1, 2, 3 and 4 repeats of 6 base GG(GT)1GG (SEQ ID NO:25) (Table 26).
    TABLE 26
    % inhibition of Jurkat human leukemia
    T cell proliferation
    SEQUENCE % Inhibition
    GGGTGG-GG(GT)1GG 60
    SEQ ID NO:25-(6 bases)
    GGGTGGGGGTGG-[GG(GT)1GG]2 18
    SEQ ID NO:68-(12 bases)
    GGGTGGGGGTGGGGGTGG-[GG(GT)1GG]3 5
    SEQ ID NO:69-(18 bases)
    GGGTGGGGGTGGGGGTGGGGGTGG-[GG(GT)1GG]4 13
    SEQ ID NO:70-(24 bases)

    As shown in Table 26, inhibition of Jurkat T cell proliferation was 60% with 6 base GT SEQ. ID NO:25 and decreased with 12 base GT SEQ ID NO:68, 18 base GT SEQ ID NO:69 and 24 base GT SEQ ID NO:70. The melting temperature (Tm) of 6 base GT SEQ ID NO:25 was 2.5° C. and increased to 56.8° C. with GT SEQ ID NO:68, to 76.3° C. with GT SEQ ID NO:69 and to 86.3° C. with GT SEQ ID NO:70.
  • EXAMPLE 20
  • Inhibition of Proliferation by Bacillus Calmette-Guerin (BCG) Derived Sequences
  • BCG derived sequences are reported to inhibit tumor growth in vivo (Kataoka et al. Jpn. J. Cancer Res. 83:244, 1992). In addition, A-2 (SEQ ID NO:72) and BCG A-4 (SEQ ID NO:74), when promixed with IMC cells and injected into CDF-1 mice, are reported to inhibit IMC tumor growth by 88% and 37% respectively.
  • Jurkat human leukemia T cells were incubated with 45 base sequences derived from BCG (Table 27).
    TABLE 27
    % inhibition of Jurkat human leukemia T
    cell proliferation
    SEQUENCE % INHIBITION
    BCG A-1
    AAAGAGGGGCATGACCCGGTGC 6
    GGGGGTTCTTGCACTCGGCATAG
    SEQ ID NO:71 (45 bases)
    BCG A-2
    AAAAGAAGTGGGGTGCCCCCAC 19
    GATCACCAACGATGGTGTGTCCA
    SEQ ID NO:72-(45 bases)
    BCG A-3
    TCCATCGCCAAGGAGATCGAGC 24
    TGGAGGATCCGTACGAGAAGATC
    SEQ ID NO:73-(45 bases)
    BCG A-4
    ACCGATGACGTCGCCGGTGACG 9
    GCAACACGACGGCCACCGTGCTG
    SEQ ID NO:74-(45 bases)
    BCG A-6
    ACGAGACCACCATCGTCGAGGG 21
    CGCCGGTGACACCGACGCCATCG
    SEQ ID NO:75-(45 bases)
    BCG A-7
    GCCGAGAAGGTGCGCAACCTGC 4
    CGGCTGGCCACGGACTGAACGCT
    SEQ ID NO:76-(45 bases)
    BCG M-3
    ACGCCGACGTCGTCTGTGGTGG 22
    GGTGTCTACCGCCAACGCGACGG
    SEQ ID NO:77-(45 bases)
    BCG ALPHA-1
    CGACTACAACGGCTGGGATATC 10
    AACACCCCGGCGTTCGAGTGGTA
    SEQ ID NO:78-(45 bases)

    As shown in Table 27, BCG derived sequences inhibited Jurkat T cell proliferation ≦24%.
  • EXAMPLE 21
  • Induction of Cell Cycle Arrest
  • Cell cycle stage was determined using a CYCLETES™ PLUS DNA commercial kit (Becton Dickinson). Briefly, nuclei from cells were obtained by dissolving the cell membrane in a nonionic detergent, eliminating the cell cytoskeleton and nuclear proteins with trypsin, digesting the cellular RNA with RNase and stabilizing the nuclear chromatin with spermine. Propidium iodide was added to the cell nuclei and their fluorescence was analyzed in a flow cytometer equipped with electronic doublet discrimination capability (FACSCalibur, Becton Dickinson, San Jose, Calif.). Accumulation of cells in G0/G1, early S (SE), mid S (SM), late S (SL) or G2/M phases of the cell cycle was analyzed using MODFIT LT software (Verity Software House Inc., Topsham, Mass.).
  • Exponentially growing Jurkat human leukemia T cells (Table 28) and MCF-7 human breast cancer cells (5×105 cells/ml) (Table 29) were incubated for 24 h with 2, 6, 15, 18, 27 and 45 base sequences containing A, C, G and T. The cells were collected and centrifuged and cell cycle stage was determined.
    TABLE 28
    Induction of cell cycle arrest in Jurkat
    T human leukemia cells
    % of cells in phase:
    G0/G1 SE SM SL G2/M Arrest*
    Untreated cells 31.4 19.1 14.3 11.6 23.6 None
    GG(GT)1GG 28.5 46.3 14.6 10.7 0.0 End SE
    SEQ ID NO:25-(6 bases)
    TT(GT)1TT 32.6 11.5 12.8 10.7 32.4 END G2/M
    SEQ ID NO:26-(6 bases) (weak)
    GT(GT)1GT 30.8 41.9 16.8 10.2 0.3 End SE
    SEQ ID NO:10-(6 bases)
    AG(GT)1GA 35.2 29.1 10.4 8.2 17.1 Mid SE
    SEQ ID NO:31-(6 bases)
    GG(AA)1GG 48.0 19.8 8.5 5.8 34.1 End
    SEQ ID NO:42-(6 bases) G0/G1
    GG(CC)1GG 26.5 21.3 22.8 10.7 18.7 End SM
    SEQ ID NO:43 (6 bases) (weak)
    GG(GT)1GG 34.9 14.8 15.0 10.6 24.7 None
    SEQ ID NO:25-(6 bases
    phosphorothioate)
    (G1T)13G 40.6 35.6 14.2 9.3 0.3 End SE
    SEQ ID NO:1-(27 bases)
    (G1T)13G 33.7 17.6 13.2 11.0 24.5 None
    SEQ ID NO:1-(27 bases
    phosphorothioate)
    (G3T)6G3 34.3 15.5 13.6 10.3 26.4 None
    SEQ ID NO:2-(27 bases)
    (G5T)4G3 40.5 13.3 12.9 9.7 23.6 None
    SEQ ID NO:3-(27 bases)
    (G7T)3G3 36.5 16.3 13.8 11.1 22.3 None
    SEQ ID NO:4-(27 bases)
    AACCACAAGCCCAAC 39.6 13.5 12.8 9.5 24.6 None
    SEQ ID NO:67-(15 bases)
    TT(AG)1GG 24.6 37.2 19.5 5.9 12.8 Mid SM
    SEQ ID NO:49-(6 bases)
    (GT)9 24.2 26.7 24.0 8.7 16.4 Mid SM
    SEQ ID NO:18-(18 bases)
    BCG A-1 19.8 31.7 22.5 14.0 12.0 Mid SM
    SEQ ID NO:71-(45 bases)
    BGC A-3 32.3 20.2 14.1 12.0 21.4 None
    SEQ ID NO:73-(45 bases)
    TG 23.1 52.3 14.8 9.8 0.0 End SE
    SEQ ID NO:51-(2 bases)
  • As shown in Table 28, in Jurkat T cells, 2, 6 and 27 base GT sequences induced arrest in the SE phase of the cell cycle, 6 base CG and AGT, 18 base GT and 45 base BCG A-1 sequences induced arrest in the SM phase of the cell cycle and a 6 base AG sequence induced arrest in the G0/G1 phase of the cell cycle.
    TABLE 29
    Induction of cell cycle arrest in MCF-7 human
    breast cancer cells
    % cells in phase:
    G0/G1 SE SM SL G2/M Arrest*
    Untreated cells 23.6 14.4 10.8 11.1 40.1 None
    GG(GT)1GG 21.9 27.6 22.2 10.9 17.4 End SM
    SEQ ID NO:25-(6 bases)
    TT(AG)1GG 20.0 18.6 25.7 20.7 15.0 Mid SM
    SEQ ID NO:49-(6 bases)
    (GT)9 25.3 31.6 16.9 10.5 15.7 Mid SM
    SEQ ID NO:18-(18 bases)
    TG 17.2 36.4 13.4 14.1 17.9 End SE
    SEQ ID NO:51-(2 bases)

    As shown in Table 29, in MCF-7 cells, 2 and 6 base GT sequences induced arrest in the SE phase of the cell cycle, a 6 base AGT sequence and an 18 base GT sequence induced arrest in the SM phase of the cell cycle.
  • EXAMPLE 22
  • Induction of Cell Cycle Arrest by GT SEQ ID NO:25, AC SEQ ID NO:79 and GT SEQ ID NO:25+AC SEQ ID NO:79
  • Jurkat human cell leukemia T cells (1×106 cells/ml) were incubated for 24 h with 6 base GT SEQ ID NO:25, conplementary 6 base AC SEQ ID NO:79 and 6 base GT SEQ ID NO:25+6 base AC SEQ ID NO:79. GT SEQ ID NO:25 and AC SEQ ID NO:79 were hybridized by mixing the oligonucleotides (1:1) and heating for 10 minutes at 65° C. As controls, GT SEQ ID NO:25 and AC SEQ ID NO:79 were heated for 10 minutes at 65° C. (Table 30).
    TABLE 30
    Induction of cell cycle arrest in Jurkat human
    leukemia T cells
    % cells in phase:
    G0/G1 SE SM SL G2/M Arrest
    Untreated cells 31.7 15.2 13.7 14.0 25.4 None
    GG(GT)1GG 28.0 45.8 14.0 11.3 0.9 End SE
    SEQ ID NO:25-(6 bases)
    CC(AC)1CC 36.0 10.4 13.4 9.7 30.5 None
    SEQ ID NO:79-(6 bases)
    GT(GT)1GT + CC(AC)1CC 35.0 13.0 10.1 8.7 33.2 None
    SEQ NO:25 + SEQ NO:79-
    (12 bases)

    As shown in Table 30, 6 base GT SEQ ID NO:25 induced arrest at the end of the SE phase of the cell cycle, whereas the complementary 6 base AC SEQ ID NO:79 had no effect on the cell cycle. Hybridization of GT SEQ ID NO:25 and AC SEQ ID NO:79 neutralized GT SEQ ID NO:25 induction of cell cycle arrest. These data demonstrate that to be effective, the sequences of the present invention must be single stranded.
  • EXAMPLE 23
  • Induction of Apoptosis
  • Redistribution of plasma membrane phosphatidylserine and release of nuclear matrix protein (NuMA) are characterisrics of cells undergoing apoptosis (Martin et al. J. Exp. Med., 182:1545, 1995; Miller et al. Biotechniques, 15:1042, 1993).
  • The redistribution of phosphatidylserine in the plasma membrane during apoptosis was measured by flow cytometry using FITC-conjugated annexin V (BD Pharmingen, San Diego, Calif.). NuMA release into the supernatant was determined using a commercial ELISA kit (Oncogen/Calbiochem, Cambridge, Mass.).
  • Jurkat human leukemia T cells were incubated with 50 μM of 3, 4, 5, 6 and 7 GT base sequences, a 5 base ACGT sequence, 6 base AG, GG, AGT and CGT sequences and a 7 base GG sequence. Table 31 shows % of cells in apoptosis (positive for phosphatidyl-serine/annexin V staining (PS/A-V)) and % NuMA released from the cells.
    TABLE 31
    Induction of apoptosis in Jurkat T cell
    leukemia cells
    % of cells in % NuMA
    apoptosis released
    (positive for (treated vs
    PS/A-V untreated
    SEQUENCE staining) cells)
    Untreated cells 4 0
    GG(GT)1GG 27 69
    SEQ ID NO:2-(6 bases)
    GG(GA)1GG 27 74
    SEQ ID NO:45-(6 bases)
    GG(GC)1GG 16 11
    SEQ ID NO:46-(6 bases)
    GG(GG)1GG 5 0
    SEQ ID NO:44-(6 bases)
    AA(GT)1AA 20 56
    SEQ ID NO:27-(6 bases)
    CC(GT)1CC 6 0
    SEQ ID NO:28-(6 bases)
    TT(GT)1TT 14 23
    SEQ ID NO:26-(6 bases)
    GT(GT)1GT 33 90
    SEQ ID NO:10-(6 bases)
    GG(GT)1 21 64
    SEQ ID NO:78-(4 bases)
    (GT)1GG 24 60
    SEQ ID NO:52-(4 bases)
    G(GT)1G 24 112
    SEQ ID NO:56-(4 bases)
    (GT)1G 21 97
    SEQ ID NO:8-(3 bases)
    T(GT)1 10 35
    SEQ ID NO:7-(3 bases)
    GG(GT)1G 25 92
    SEQ ID NO:6-(5 bases)
    G(GT)1GG 25 120
    SEQ ID NO:60-(5 bases)
    GG(GG)1GGG 12 26
    SEQ ID NO:63-(7 bases)
    GGG(GT)1GG 30 123
    SEQ ID NO:62-(7 bases)
    CG(GT)1A 6 9
    SEQ ID NO:80-(5 bases)
  • As shown in Table 31, 3, 4, 5, and 6 base GT, AG, CG and AGT sequences induced apoptosis of Jurkat T cells. Five base ACGT and 6 base CGT and GG sequences did not induce apoptosis of Jurkat T cells.
  • EXAMPLE 24
  • Increase in Iintracellular Calcium (Ca2+)i
  • Increases in intracellular calcium (Ca2+)i are reported to be associated with apoptosis induction (Lam et al. Mol. Endocrinol. 7:686, 1993). (Ca2+)i was followed using the fluorescent probe Fluo-3-AM (Cell Permaant, Molecular Probes, Inc., Eugene, Oreg.). An increase in Fluo-3-AM fluorescence is indicative of an increase in (Ca2+)i.
  • Jurkat human leukemia T cells were incubated for 24 h with 6 base GT SEQ. NO:25. Cells were collected by centrifugation, suspended in PBS containing 1% FBS and loaded with 10 μM Fluo-3-AM for 1 h at 37° C. Cell fluorescence was measured at 488 nm excitation and 530 nm emission (FL1 detector). Data were analyzed on a FACSCALIBUR using the program CellQUEST (Becton Dickinson).
  • As shown in FIG. 1, incubation of Jurkat T cells with 6 base GT SEQ ID NO:25 caused an 88% increase in cell fluorescence, indicative of an increase in (Ca2+)i.
  • EXAMPLE 25
  • Induction of Apoptosis
  • Apoptosis can be initiated by ligands that bind to cell surface receptors including, but not limited to, Fas (CD95) and tumor necrosis factor (TNF). Fas binding to Fas Ligand and TNF binding to TNF Receptor 1 initiate intracellular signaling resulting in the activation of cysteine aspartyl proteases (caspases). Caspases initiate the lethal proteolytic cascade of apoptosis execution associated with nuclear DNA-fragmentation, release of nuclear matrix proteins (NuMA) and loss of cell substrate contact.
  • Jurkat human leukemia T cells (1×106/ml) were incubated with 6 and 27 GT base sequences (Table 32). NuMA was determined as in Example 23.
    TABLE 32
    % NuMA release from Jurkat human leukemia
    T cells
    % NUMA
    SEQUENCE RELEASED
    GTGTGTGTGTGTGTGTGTGTGTGTGTG-(G1T)13G 22
    SEQ ID NO:1-(27 bases)
    GGGTGGGTGGGTGGGTGGGTGGGTGGG-(G3T)6G3 49
    SEQ ID NO:2-(27 bases)
    GGGGGTGGGGGTGGGGGTGGGGGTGGG-(G5T)4G3 139
    SEQ ID NO:3-(27 bases)
    GGGGGGGTGGGGGGGTGGGGGGGTGGG-(G7T)3G3 90
    SEQ ID NO:4-(27 bases)
    GGGTGG-GG(GT)1GG 269
    SEQ ID NO:25-(6 bases)

    As shown in Table 32, % NuMA release with 6 base GT SEQ ID NO:25 was greater than % NuMA release with 27 base GT SEQ ID NOs:1, 2, 3 and 4.
  • EXAMPLE 26
  • Induction of Apoptosis by 6 Base GT SEQ ID NO:25 and 6 Base AC SEQ ID NO:79
  • Jurkat human cell leukemia T cells were incubated for 24 h with 6 base GT SEQ ID NO:25, complementary 6 base AC SEQ ID NO:79, and 6 base GT SEQ ID NO:25+6 base AC SEQ ID NO:79. GT SEQ ID NO:25 and AC SEQ ID NO:79 were hybridized by mixing the sequences (1:1) and heating for 10 minutes at 65° C. As controls, GT SEQ ID NO:25 and AC SEQ ID NO:79 were heated for 10 minutes at 65° C. (Table 33). Apoptosis was evaluated as in Example 23.
    TABLE 33
    Induction of apoptosis in Jurkat human
    leukemia T cells
    % cells in % NuMA
    apoptosis released
    (positive for (untreated vs
    PS/A-V treated
    staining) cells)
    Untreated cells 4 0
    GG(GT)1GG 27 69
    SEQ ID NO:25-(6 bases)
    CC(AC)1CC 6 9
    SEQ ID NO:79-(6 bases)
    GT(GT)1GT + 5 9
    CC(AC)1CC
    SEQ ID NO:25-(6 bases) +
    SEQ ID NO:79-(6 bases)

    As shown in Table 33, 6 base GT SEQ ID NO:25 induced apoptosis of Jurkat T cells, whereas the complementary AC SEQ ID NO:79 had no effect on apoptosis. Moreover, hybridization of GT SEQ ID NO:25 and AC SEQ ID NO:79 neutralized GT SEQ ID NO:25's induction of apoptosis. These data demonstrate that to be effective, the sequences of the present invention must be single stranded.
  • EXAMPLE 27
  • Inhibition of Proliferation, Cell Cycle Arrest and Induction of Apoptosis by GT-Rich and AC-Rich Sequences Derived from Mycobacterium phlei
  • Jurkat human leukemia T cells were incubated with GT-rich or AC-rich sequences derived from the murA gene of Mycobacterium phlei (GenBank: Accession Number X99776). Inhibition of proliferation was measured by the reduction of MTT as in Example 3, cell cycle arrest was detected by flow cytometry using propidium iodide as in Example 21 and apoptosis was evaluated by flow cytometry using annexin-V-FITC as in Example 23.
    TABLE 34
    Inhibition of proliferation, cell cycle
    arrest and induction of apoptosis in
    Jurkat cells
    % at cell
    % inhibition cells in cycle
    SEQUENCE (proliferation) apoptosis arrest
    AACCACAAGCCCAAC 4 0 No
    SEQ ID NO:67-
    (15 bases)
    GTGTGTTTGGT 22 23 G0/G1
    SEQ ID NO:81-
    (11 bases)
    GGTTTTGTTTG 20 25 End SE
    SEQ ID NO:82-
    (11 bases)
    TTGTTTTTTTTG 21 16 SM
    SEQ ID NO:83-
    (11 bases)

    As shown in Table 34, SEQ ID NOs:81, 82 and 83, rich in GT, inhibited proliferation of, induced cell cycle arrest in and induced apoptosis of Jurkat T cells, whereas SEQ ID NO:15, rich in AC, did not inhibit proliferation of, induce cell cycle arrest in or induce apoptosis of Jurkat T cells.
  • EXAMPLE 28
  • Modulation of Caspase Activation by GT Sequences
  • Caspases recognize 3 major peptide substrate sequences: 1) Tyr-Val-Ala-Asp (YVAD, caspase-1, -4 and -5) (SEQ ID NO:84); 2) Asp-Glu-Val-Asp (DEVD, caspase-2, -3 and -7) (SEQ ID NO:85); and, 3) Ile-(Leu)-Glu-X-Asp (I(L)EXD; caspase-8 and -10) (SEQ ID NO:86) (Thomberry et al. J. Biol. Chem. 272:17907, 1997). Sequence recognition in a protein target results in a limited and specific proteolysis of the target as, in a first example, the modulation of caspase 7 activation by caspase 3 or, as in a second example, the degradation of structural protein targets including, but not limited, to lamins or, as in a third example, the activation of enzymes including, but not limited to, PARP.
  • NH2—XXXD-COO-GT sequence constructs are generated by chemical conjugation of a chemically protected GT sequence or of a chemically protected AC sequence to a chemically protected peptide selected from the group consisting of NH2—YVAD-COOH (SEQ ID NO:84), NH2-DEVD-COOH (SEQ ID NO:85) and NH2-IEGD-COOH(SEQ ID NO:87) using an oligonucleotide synthesized with a 5′-C2 amide spacer arm and standard amide-carboxyl water soluble carbohexiimide conjugation techniques (Guy et al. J. Chromatography. B. Biomed. Sci. Appl. 706:149, 1998). Reactive carboxylate and reactive amine groups are deprotected subsequent to conjugation,
  • Peptide-GT (hereinafter, PGT) sequence constructs including, but not limited to, NH2—YVAD-COO-GT; NH2-DEVD-COO-GT; and, NH2—I(L)EXD-COO-GT are cleaved at the carboxylate function between D and the GT sequence by enzymes including, but not limited to, caspases, cancer metastasisassociated enzymes, collagenase and metalloproteinases. Such cleavage results in the release of the caspase-activating GT sequence from the PGT. The resulting increase in intracellular caspase activity can, for example, enhance the therapeutic effect of chemotherapeutic agents in multidrug resistant cancer cells or the immune response to weakly antigenic stimuli.
  • To determine caspase activation, control and treated cells are washed, fixed, permeabilized and incubated with an FITC-conjugated antibody that recognizes the active form of the caspase (Pharmingen, San Diego, Calif.) using the conditions recommended by the manufacturer. Fluorescence associated with active caspase 3 is analyzed by flow cytometry on a FACSCALIBUR using the program CellQUEST (Becton Dickinson). Alternatively, caspase activation is determined colorimetrically using an assay based on the cleavage of a caspase-specific peptide conjugated to the color reporter molecule p-nitroanilide, which can be quantitated spectrophotometrically at a wavelength of 405 nm.
  • EXAMPLE 29
  • Activation of Caspase 3 by GT SEQ ID NOs: 66, 81, 82 and 83 and by AGC sequence SEQ ID NO:67
  • Jurkat T cell leukemia cells were incubated for 72 h with 33 base GT SEQ ID NO:66; 11 base GT SEQ ID NO:81 (bases 1-11 of GT SEQ ID NO:66), 11 base GT SEQ ID NO:82 (bases 12-22 of GT SEQ ID NO:66), 11 base GT SEQ ID NO:83 (bases 23-33 of GT SEQ ID NO:66) and 15 base ACG SEQ ID NO:67. Active caspase 3 (17-22 kDa) was determined using FITC conjugated antibody (Clone: C92-605) as in Example 28.
  • As shown in FIG. 2A, 33 base GT SEQ ID NO:66 and 11 base GT SEQ ID NOs:81, 82 and 83 each induced processing of inactive pro-caspase 3 to active caspase 3, whereas 15 base ACG SEQ ID NO:67 did not induce processing of inactive pro-caspase 3 to active caspase 3.
  • Caspase 3 activation also was determined colorimetrically as in Example 28. As As shown in FIG. 2B, caspase 3 activity in 33 base GT SEQ ID NO:66 and 11 base GT SEQ ID NOs:81, 82 and 83 treated cells was 105%, 77%, 100% and 60% greater than that in control cells, whereas in 15 base ACG SEQ ID NO:67 treated cells caspase 3 activation was approximately the same as in control cells.
  • EXAMPLE 30
  • Activation of Caspase 3 Activity by 6 Base GT SEQ ID NO:25
  • Jurkat T cell leukemia cells were incubated for 72 h with 6 base GT SEQ ID NO:25. Caspase 3 activation was determined colorimetrically as in Example 28. As shown in FIG. 3A, caspase 3 activation in 6 base GT SEQ ID NO:25 treated cells was 323% greater than in control cells.
  • EXAMPLE 31
  • Activation of Caspase-7 and PARP Cleavage by GT SEQ ID NO:25
  • Jurkat T cell leukemia cells were incubated for 72 h with 6 base GT SEQ ID NO:25. The cells were washed 3× with PBS, lysed with 10 mM HEPES, pH 7.5 containing 5 mM MgCl2, 1 mM dithiothreitol, 1.5 nM aprotinin, 10 mM leupeptin and 2.5 μm Na orthovanadate, and the protein content of the lysate was determined (Bradford J. Anal. Biochem. 72:248, 1976).
  • Activated caspase 7 and PARP cleavage were detected by Western blot analysis. Lysate was mixed with Laemmli buffer (Laemmli U. Nature 15:680, 1970), shaken, and heated at 100° C. for 4 min. Fifty μg of protein was added to each lane and the proteins were separated by electrophoresis in a 10% (caspase) or a 17% (PARP) sodium dodecyl sulfate-polyacrylamide gels (SDS-PAGE) at a constant voltage of 100 V for about 1.5 h. The separated proteins were electroblotted onto a PVDF membrane. Equal protein loading was monitored by Ponceau red staining of the membrane.
  • The membrane was blocked overnight at 4° C. with a buffer containing 1% Tris-buffered saline (2 mM Tris-HCl, 13.7 mM NaCl, pH 7.6, and 0.1% polyethylenesorbitan monolaurate (TWEEN 20) (TBST) +5% non-fat dry milk. The membrane was washed and was incubated for 1 h at RT with a mouse monoclonal IgG anti-caspase 7 antibody (Pharmingen) (diluted 1:1000 in TBST+1% BSA) or with a mouse monoclonal IgG anti-PARP antibody (diluted 1:1000 in TBST+1% BSA) (Pharmingen). IgG bound to caspase 7 or to PARP was detected with sheep anti-mouse IgG conjugated to horseradish peroxidase (diluted 1:1000 in TBST+5% non-fat dry milk) (Pharmingen). Blots were developed using an enhanced chemiluminescence detection system (ECL, Amersham, Corp., Amersham, UK).
  • As shown in FIG. 3B, 6 base GT SEQ ID NO:25, induced processing of inactive pro-caspase 7 (30 kDa) to active caspase 7 (19-20 kDa) and active PARP to its inactive 85 kDa degradation product.
  • EXAMPLE 32
  • Positive Feedback Amplification of Caspase Activation
  • Jurkat human leukemia T cells are incubated for 72 h with NH2—YVAD-CO-GG(GT)GG, NH2-DEVD-CO-GG(GT)GG, NH2—EGD-COO-GG(GT)GG, NH2-YVAD-CO-AACCACAAGCCCAAC, NH2-DVED-CO-AACCACAAGCCCAAC and NH2—IEGD-CO-AACCACAAGCCCAAC. Caspase 3 and caspase 7 activation are determined.
  • NH2—YVAD-CO-GT, NH2-DEVD-CO-GT and NH2—IEGD-COO-GT each induce processing of inactive caspase 3 to active caspase 3 and of inactive caspase 7 to active caspase 7, whereas NH2—YVAD-CO-ACG, NH2-DVED-CO-ACG and NH2—IEGD-CO-ACG do not induce processing of inactive pro-caspase 3 to active caspase 3 or of inactive caspase 7 to active caspase 7.
  • Although not wanting to be bound by the following hypothesis, it is thought that basal caspase activity within caspase containing cells mediates the release of caspase activating GT sequences from NH2—XXXD-COO-GT constructs by proteolysis/hydrolysis. This results in positive amplification of caspase activity (increased levels of caspase 3 and caspase 7) within the cells by the released caspase-activating GT sequences.
  • EXAMPLE 33
  • Induction of Cytokine Production
  • Unless stated otherwise, 1×106 cells were incubated with 100 μg/ml of each of the sequences tested for 48 h at 37° C. in 5% CO2. Production of cytokines IL-6, IL-10, IL-12, IL-1beta and TNF-alpha was determined in pg/ml in 100 μl of culture supernatant using the appropriate commercial ELISA (BioSource, Camarillo Calif.). The IL-12 ELISA measures both IL-12 p70 complex and free p40 subunit. Results are expressed as the “fold” (×) increase in cytokine production by treated cells compared to control cells.
  • THP-1 human acute monocytic leukemia cells were incubated with 2, 3, 6, 9, 12, 14, 15 and 18 base GT sequences and production of the cyotkine IL-6 was determined (Table 35).
    TABLE 35
    Cytokine production by THP-1 human acute
    monocytic leukemia cells
    SEQUENCE ↑ IL-6
    TG-(TG)1T 2.7x
    SEQ ID NO:7-(3 bases)
    TG-(TG)1 2.9x
    SEQ ID NO:51-(2 bases)
    TGTGTG-(TG)3 4.0x
    SEQ ID NO:9-(6 bases)
    GTGTGT-(GT)3 5.0x
    SEQ ID NO:10-(6 bases)
    TGTGTGTG-(TG)4T 5.4x
    SEQ ID NO:11-(9 bases)
    GTGTGTGTG-(GT)4G 5.4x
    SEQ ID NO:12-(9 bases)
    TGTGTGTGTGT-(TG)6 5.7x
    SEQ ID NO:13-(12 bases)
    GTGTGTGTGTGT-(GT)6 1.3x
    SEQ ID NO:14-(12 bases)
    TGTGTGTGTGTGT-(TG)7 1.0x
    SEQ ID NO:15-(14 bases)
    GTGTGTGTGTGTGTG-(GT)7G 2.6x
    SEQ ID NO:16-(15 bases)
    TGTGTGTGTGTGTGTGT-(TG)9 2.2x
    SEQ ID NO:17-(18 bases)
    GTGTGTGTGTGTGTGTGT-(GT)9 2.8x
    SEQ ID NO:18-(18 bases)
  • As shown in Table 35, 2, 3, 6, 9, 12, 14, 15 and 18 base GT sequences increased THP-1 cell production of the cytokine IL-6.
  • THP-1 human acute monocytic leukemia cells were incubated with 6 base GT, AG, CG, GG, AGT and CGT sequences and production of the cytokines IL-12 and IL-6 was determined (Table 36).
    TABLE 36
    Cytokine production by THP-1 human acute
    monocytic leukemia cells
    SEQUENCE ↑ IL-12 ↑ IL-6
    TGTGTG-(TG)3 SEQ ID NO:9 1.8x 4.0x
    GTGTGT-(GT)3 SEQ ID NO:10 2.2x 5.0x
    TTTGTT-TT(TG)1TT SEQ ID NO:23 3.5x 4.9x
    GGTGGG-GG(TG)1GG SEQ ID NO:24 3.7x 6.9x
    GGGTGG-GG(GT)1GG SEQ ID NO:25 2.3x 3.1x
    TTGTTT-TT(GT)1TT SEQ ID NO:26 3.5x 5.3
    AAGTAA-AA(GT)1AA SEQ ID NO:27 6.0x 12.8x
    CCGTCC-CC(GT)1CC SEQ ID NO:28 3.8x 12.6x
    TGGTTG-TG(GT)1TG SEQ ID NO:29 4.1x 10.5x
    ATGTAT-AT(GT)1AT SEQ ID NO:30 4.8x 9.8x
    AGGTGA-AG(GT)1GA SEQ ID NO:31 1.9x 4.9x
    GAGTGA-GA(GT)1GA SEQ ID NO:32 1.8x 5.8x
    GGGTCT-GG(GT)1CT SEQ ID NO:33 1.2x 3.1x
    CCGTGG-CC(GT)1GG SEQ ID NO:34 0.0x 10.8x
    GGGTCC-GG(GT)1CC SEQ ID NO:35 1.9x 21.3x
    CTGTCT-CT(GT)1CT SEQ ID NO:36 2.0x 15.9x
    TCGTTC-TC(GT)1TC SEQ ID NO:37 2.2x 12.9x
    CGGTGC-CG(GT)1GC SEQ ID NO:38 0.2x 6.9x
    TTGTG-TT(GT)1GG SEQ ID NO:39 0.0x 6.6x
    GGGTT-GG(GT)1TT SEQ ID NO:40 −1.2x 14.0x
    GGTTGG-GG(TT)1GG SEQ ID NO:41 3.3x 16.0x
    GGAAG-GG(AA)1G SEQ ID NO:42 4.1x 29.2x
    GGCCGG-GG(CC)GG SEQ ID NO:43 3.1x 17.1x
    GGGGGG-GG(GG)1GG SEQ ID NO:44 0.0x 15.1x
    GGGAGG-GG(GA)1GG SEQ ID NO:45 −1.6x 23.2x
    GGGCGG-GG(GC)1GG SEQ ID NO:46 2.3x 9.8x
    TTAGGG-TT(AG)1GG SEQ ID NO:49 2.0x 6.7x
  • As shown in Table 36, 6 base GT, AG, CG, GG, AGT and CGT sequences increased THP-1 cell production of the cytokines IL-12 and IL-6.
  • Table 37 summarizes the induction of IL12 and IL-6 synthesis by 6 base sequences.
    TABLE 37
    IL-6 and IL-12 synthesis induced by 6 base sequences
    Fold IL-12 synthesis IL-6 synthesis
    increase SEQ ID NOs: SEQ ID NOs:
      ≦2.0 9, 31, 32, 33, 34, 35, 36, 38,
    39, 40, 44, 45, 49
    >2.0 and 10, 23, 24, 25, 26, 27, 28, 29, 9, 10, 23, 24, 25, 26, 30, 31,
    ≦ 10.0 30, 37, 40, 41, 42, 43, 44, 45 32, 33, 38, 39, 46, 49
    >10.0 25, 27, 29, 34, 35, 36, 37,
    40, 41, 42, 43, 44, 45
  • BCG derived sequences A-3 (SEQ ID NO:73), A-4 (SEQ ID NO:74), A-6 (SEQ ID NO:75), A-7 (SEQ ID NO:76), M3 (SEQ ID NO:77) and Alpha 1 (SEQ ID NO:78) are reported to activate NK cells in vivo (Kataoka et al. Jpn. J. Cancer Res. 83:244, 1992). THP-1 human acute monocytic leukemia cells were incubated with 45 base BCG-derived sequences and production of the cytokines IL-12 and IL-6 was determined (Table 38).
    TABLE 38
    Cytokine production by THP-1 human acute
    monocytic leukemia cells
    SEQUENCE ↑ IL-12 ↑ IL-6
    BCG A-1
    AAAGAGGGGCATGACCCGGTGC 1.9x 2.6x
    GGGGCTTCTTGCACTCGGCATAG
    SEQ ID NO:69-(45 bases)
    BCG A-2
    AAAAGAAGTGGGGTGCCCCCAC 1.6x 3.9x
    GATCACCAACGATGGTGTGTCCA
    SEQ ID NO:70-(45 bases)
    BCG A-3
    TCCATCGCCAAGGAGATCGAGC 1.7x 2.5x
    TGGAGGATCCGTACGAGAAGATC
    SEQ ID NO:71-(45 bases)
    BCG A-4
    ACCGATGACGTCGCCGGTGACG 0.9x 1.8x
    GCAACACGACGGCCACCGTGCTG
    SEQ ID NO:72-(45 bases)
    BCG A-6
    ACGAGACCACCATCGTCGAGGG 2.1x 3.9x
    CGCCGGTGACACCGACGCCATCG
    SEQ ID NO:73-(45 bases)
    BCG A-7
    GCCGAGAAGGTGCGCAACCTGC 0.5x N.D.
    CGGCTGGCCACGGACTGAACGCT
    SEQ ID NO:74-(45 bases)
    BCG M-3
    ACGCCGACGTCGTCTGTGGTGG 1.6x 2.5x
    GGTGTCTACCGCCAACGCGACGG
    SEQ ID NO:75-(45 bases)
    BCG ALPHA-1
    CGACTACAACGGCTGGGATATC 1.1x 2.1x
    AACACCCCGGCGTTCGAGTGGTA
    SEQ ID NO:76-(45 bases)
  • As shown in Table 38, 45 base BCG derived sequences minimally increased THP-1 cell production of IL-12 and IL-6.
  • EXAMPLE 34
  • Induction of IL-12 Production by Phosphodiester and Phosphorotothioate Sequences
  • THP-1 human acute monocytic leukemia cells were incubated with 6 base GT sequences, having either an oxygen (phosphodiester) or a sulfur (phosphorothioate) as the nonbridging atom on the phosphate groups and production of the cytokine IL-12 was determined (Table 39).
    TABLE 39
    IL-12 production by THP-1 human acute monocytic
    leukemia cells
    INCREASE
    SEQUENCE PHOSPHODIESTER PHOSPHOROTHIOATE
    TGTGTG-(TG)3 (6 bases) 1.8x −0.1x
    SEQ ID NO:9 phosphodiester;
    phosphorothioate
    GTGTGT-(GT)3 (6 bases) 2.2x −0.2x
    SEQ ID NO:10 phosphodiester;
    phosphorothioate
    TTTGT-TT(TG)1TT (6 bases) 3.5x 0.1x
    SEQ ID NO:23 phosphodiester;
    phosphorothioate
    GGTGGG-GG(TG)1GG (6 bases) 3.7x −0.1x
    SEQ ID NO:24 phosphodiester;
    phosphorothioate
    GGGTGG-GG(GT)1GG (6 bases) 2.0x 0.0x
    SEQ ID NO:25 phosphodiester;
    phosphorothioate
    TTGTTT-TT(GT)1TT (6 bases) 3.8x −0.1x
    SEQ ID NO:26 phosphodiester;
    phosphorothioate

    As shown in Table 39, substitution of a sulfur atom (phosphorothioate) for a nonbridging oxygen atom (phosphodiester) on the phosphate groups resulted in a significant decrease in THP-1 cell production of IL-12.
  • THP-1 human acute monocytic leukemia cells were incubated with 6 base GT SEQ ID NO:25, having either an oxygen (phosphodiester) or a sulfur (phosphorothioate) as the nonbridging atom on the phosphate groups and production of the cytokine IL-12 was determined (Table 40).
    TABLE 40
    IL-12 production by THP-1 human acute
    monocytic leukemia cells
    SEQUENCE* INCREASE
    GoGoGoToGoGo-GoGo(GoTo)1GoGo; 2.0
    (oxygen atom: base 1 to 6)
    SEQ ID NO:25-(6 bases)
    GoGoGsToGoGo-GoGo(GsTo)1GoGo; (oxygen 0.1
    atom: base 1, 2, 4, 5, 6; sulfur atom:
    base 3)
    SEQ ID NO:25-(6 bases)
    GoGoGoTsGoGo-GoGo(GoTs)1GoGo; (oxygen 0.2x
    atom: base 1, 2, 3, 5, 6; sulfur atom:
    base 4)
    SEQ ID NO:25-(6 bases)
    GoGoGsTsGoGo-GoGo(GsTs)1GoGo; (oxygen 0.5x
    atom: base 1, 2, 5, 6; sulfur atom: base
    3, 4)
    SEQ ID NO:25-(6 bases)
    GsGoGoToGoGs-GsGo(GoTo)1GoGs; (oxygen −0.1x
    atom: base 2, 3, 4, 5; sulfur atom: base
    1, 6)
    SEQ ID NO:25-(6 bases)
    GoGsGoToGsGo-GoGs(GoTo)1GsGo; (oxygen −0.1x
    atom: position 1, 3, 4, 6; sulfur atom:
    position 2, 5)
    SEQ ID NO:25-(6 bases)
    GsGsGoToGsGs-GsGs(GoTo)1GsGs; (oxygen 0
    atom: position 3, 4; sulfur atom: position
    1, 2, 5, 6)
    SEQ ID NO:25-(6 bases)
    GsGsGsTsGsGs-GsGs(GsTs)1GsGs; (sulfur 0
    atom: position 1 to 6)
    SEQ ID NO:25(6 bases)

    *Note:

    o” represents an oxygen atom and “s” represents a sulfur atom on the phosphate group
  • As shown in Table 40, substitution of a sulfur atom (phosphorothioate) for a nonbridging oxygen atom (phosphodiester) in 6 base GT SEQ ID NO:25 resulted in a significant decrease in THP-1 cell production of IL-12.
  • EXAMPLE 35
  • Stimulation of Cytokine Synthesis in Human Peripheral Blood Mononuclear Cells
  • Peripheral blood mononuclear cells (hereinafter, “PBMCs”) were isolated from 7 healthy humans by Ficoll-Hypaque (Amersham Pharmacia Biotech, Baie d'Urfée, Québec, Canada) by density gradient centrifugation of whole blood. PBMCs were incubated with 6 base GT, AGT, CGT, AG, CG and GG sequences and production of the cytokines IL-1beta, IL-6, IL-10 and IL-12 were determined.
    TABLE 41
    Cytokine production by human PBMC
    IL-1 beta IL-6 IL-10 IL-12
    fold increase: fold increase: fold increase: fold increase:
    mean +/− SD mean +/− SD mean +/− SD mean +/− SD
    SEQUENCES (range) (range) (range) (range)
    TG(TG)1TG 1.2 +/− 0.4  8.3 +/− 12.8 1.0 +/− 0.1 2.7 +/− 2.6
    SEQ ID NO:9- (0.8-1.5)  (1.2-37.0) (0.9-1.1) (1.0-6.6)
    (6 bases)
    GT(GT)1GT 2.0 +/− 1.6  9.8 +/− 14.2 1.0 +/− 0.1 4.0 +/− 6.3
    SEQ ID NO:10- (0.9-3.8)  (0.8-39.1) (0.9-1.1)  (0.9-18.1)
    (6 bases)
    TG(TG)4TG 2.4 +/− 1.9 12.1 +/− 6.5  1.2 +/− 0.4 4.4 +/− 5.3
    SEQ ID NO:13- (0.9-4.5)  (2.9-20.8) (0.9-1.9)  (1.2-15.0)
    (12 bases)
    GT(GT)4GT 1.1 +/− 0.2 2.0 +/− 1.5 1.0 +/− 0.1 2.0 +/− 1.1
    SEQ ID NO:14- (0.9-1.3) (0.9-4.9) (0.9-1.2) (0.9-2.6)
    (12 bases)
    TT(TG)1TT 1.0 +/− 0.1 11.8 +/− 9.0  1.0 +/− 0.1 1.1 +/− 0.3
    SEQ ID NO:23- (0.9-1.0)  (1.3-25.6) (0.9-1.1) (0.9-1.6)
    (6 bases)
    GG(TG)1GG 0.9 +/− 0.1 15.9 +/− 14.9 2.4 +/− 2.9 2.3 +/− 1.6
    SEQ ID NO:24 (0.9-1.0)  (0.7-37.1) (1.0-7.5) (0.9-5.5)
    (6 bases)
    GG(GT)1GG 1.0 +/− 0.1 20.9 +/− 18.0 11.6 +/− 1.2  13.2 +/− 11.5
    SEQ ID NO:25- (1.0-1.2)  (1.6-50.0)  (9.9-13.2)  (1.0-26.8)
    (6 bases)
    TT(GT)1TT 1.5 +/− 0.9 5.8 +/− 8.0 1.0 +/− 0.1 1.6 +/− 1.3
    SEQ ID NO:26- (0.9-2.5)  (0.5-21.7) (1.0-1.2) (0.8-4.5)
    (6 bases)
    AA(GT)1AA 1.3 +/− 0.5 9.6 +/− 7.3 1.0 +/− 0.1 2.4 +/− 2.2
    SEQ ID NO:27- (0.9-1.8)  (1.8-16.0) (0.9-1.1) (0.8-6.7)
    (6 bases)
    CC(GT)1CC 2.1 +/− 1.8 10.4 +/− 13.0 1.0 +/− 0.1  5.9 +/− 10.7
    SEQ ID NO:28- (0.8-4.1)  (1.4-35.8) (1.0-1.1)  (0.9-30.0)
    (6 bases)
    TG(GT)1TG 1.7 +/− 1.0  9.3 +/− 12.1 1.0 +/− 0.1 3.3 +/− 4.2
    SEQ ID NO:29- (1.1-2.8)  (0.8-33.8) (1.0-1.1)  (0.8-12.7)
    (6 bases)
    AT(GT)1AT 1.1 +/− 0.2 4.6 +/− 4.4 1.0 +/− 0.1 1.3 +/− 0.2
    SEQ ID NO:30- (1.0-1.4)  (1.6-14.4) (0.9-1.1) (1.0-1.6)
    (6 bases)
    CT(GT)1CT 1.2 +/− 0.3 5.3 +/− 3.6 1.0 +/− 0.1 1.2 +/− 0.3
    SEQ ID NO:36- (1.0-1.5)  (0.9-10.6) (0.9-1.2) (0.9-1.8)
    (6 bases)
    TC(GT)1TC 1.2 +/− 0.4 7.5 +/− 9.8 1.0 +/− 0.1 1.4 +/− 1.1
    SEQ ID NO:37- (0.9-1.6)  (0.7-27.0) (1.0-1.1) (0.9-3.8)
    (6 bases)
    GG(TT)1GG 3.2 +/− 3.0  7.8 +/− 12.8 1.0 +/− 0.1 4.9 +/− 8.6
    SEQ ID NO:41- (0.8-6.5)  (0.9-36.4) (0.9-1.1)  (0.8-24.3)
    (6 bases)
    GG(AA)1GG 3.5 +/− 3.9 11.8 +/− 5.8  1.1 +/− 0.2 3.0 +/− 2.7
    SEQ ID NO:42- (0.8-8.0)  (1.2-15.6) (0.9-1.5) (1.1-8.6)
    (6 bases)
    GG(CC)1GG 1.5 +/− 0.9 14.9 +/− 10.2 1.2 +/− 0.3 2.5 +/− 2.0
    SEQ ID NO:43- (0.8-2.5)  (1.2-29.8) (1.0-1.8) (1.0-7.0)
    (6 bases)
    GG(GG)1GG 7.1 +/− 9.4 21.0 +/− 14.7 1.7 +/− 1.6  7.0 +/− 12.5
    SEQ ID NO:44-  (0.8-17.9)  (4.6-42.0) (0.9-4.6)  (1.3-35.3)
    (6 bases)
    GG(GA)1GG 13.6 +/− 14.9 24.7 +/− 16.5 6.0 +/− 5.5 20.7 +/− 10.3
    SEQ ID NO:45-  (1.2-30.2)  (5.9-50.0)  (2.1-15.5)  (5.7-35.3)
    (6 bases)
    GG(GC)1GG 1.2 +/− 0.3 15.8 +/− 16.2 1.0 +/− 0.1 3.0 +/− 3.8
    SEQ ID NO:46- (1.0-1.5)  (1.3-37.8) (0.9-1.1)  (1.1-10.7)
    (6 bases)
  • As shown in Table 41, 6 base GT, AGT, CGT, AG, CG and GG sequences increased human PBMC cell production of the cytokines IL1beta, IL-6, IL-10 and IL-12.
  • EXAMPLE 36
  • Cytokine Synthesis by Chimpanzee Peripheral Blood Mononuclear Cells
  • PBMCs were isolated from 4 healthy chimpanzees as in Example 35. Chimpanzee PBMCs were incubated with 6 base GT, AGT, CGT, AG, CG and GG sequences and production of the cytokines IL-10, IL-12 and TNF-alpha was determined (Table 41).
    TABLE 41
    Cytokine production by chimpanzees PBMC
    IL-10 IL-12 TNF-alpha
    fold increase: fold increase: fold increase:
    mean +/− SD mean +/− SD mean +/− SD
    SEQUENCES (range) (range) (range)
    TG(TG)1TG 2.3 +/− 1.3 13.5 +/− 8.9  11.3 +/− 6.9 
    SEQ ID NO:9- (1.3-4.1)  (2.8-21.1)  (5.3-19.5)
    (6 bases)
    GT(GT)1GT 4.0 +/− 2.1 14.0 +/− 8.5  12.9 +/− 6.4 
    SEQ ID NO:1- (1.8-6.7)  (3.0-21.3)  (6.5-19.6)
    (6 bases)
    TT(TG)1TT 1.5 +/− 0.6 12.9 +/− 8.2  9.0 +/− 5.5
    SEQ ID NO:23- (1.0-2.4)  (2.7-20.1)  (4.1-14.4)
    (6 bases)
    GG(TG)1GG 2.9 +/− 1.5 14.3 +/− 9.1  11.9 +/− 7.0 
    SEQ ID NO:24- (1.3-4.8)  (3.0-22.5)  (5.8-19.8)
    (6 bases)
    GG(GT)1GG 2.5 +/− 1.5 13.5 +/− 8.4  11.7 +/− 6.7 
    SEQ ID NO:25- (1.4-4.6)  (2.8-20.8)  (5.9-19.6)
    (6 bases)
    TT(GT)1TT 1.4 +/− 0.9 12.3 +/− 8.5  7.5 +/− 4.6
    SEQ ID NO:26- (1.1-2.1)  (2.5-20.1)  (3.4-13.1)
    (6 bases)
    AA(GT)1AA 2.1 +/− 1.1 13.2 +/− 8.2  7.9 +/− 3.9
    SEQ ID NO:27 (1.1-3.7)  (2.8-19.8)  (4.6-12.0)
    (6 bases)
    CC(GT)1CC 3.8 +/− 2.8 13.3 +/− 8.4  10.3 +/− 5.7 
    SEQ ID NO:28- (1.5-7.7)  (2.8-20.4)  (5.0-15.8)
    (6 bases)
    TG(GT)1TG 3.1 +/− 2.0 13.6 +/− 8.8  12.4 +/− 7.3 
    SEQ ID NO:29- (1.6-5.9)  (2.9-21.9)  (6.1-20.8)
    (6 bases)
    AT(GT)1AT 1.4 +/− 0.4 10.7 +/− 7.0  5.9 +/− 3.3
    SEQ ID NO:30- (1.2-1.9)  (2.5-18.6)  (3.4-10.5)
    (6 bases)
    CT(GT)1CT 3.0 +/− 2.1 13.4 +/− 8.9 12.4 +/− 6.3 
    SEQ ID NO:36- (1.2-5.9)  (2.8-20.4)  (7.0-19.6)
    (6 bases)
    TC(GT)1TC 3.4 +/− 2.6 14.1 +/− 10.0 11.4 +/− 6.4 
    SEQ ID NO:37- (1.4-7.1)  (2.4-24.9)  (6.1-19.3)
    (6 bases)
    GG(TT)1GG 9.1 +/− 7.7 15.3 +/− 10.0 14.1 +/− 7.3 
    SEQ ID NO:41  (3.0-20.3)  (2.9-25.9)  (7.7-23.2)
    (6 bases)
    GG(AA)1GG 9.9 +/− 8.9 15.6 +/− 10.6 14.4 +/− 7.1 
    SEQ ID NO:42-  (2.6-22.7)  (2.6-26.6)  (8.0-22.9)
    (6 bases)
    GG(CC)1GG 13.6 +/− 9.0  15.1 +/− 10.2 14.0 +/− 6.6 
    SEQ ID NO:43-  (4.3-26.7)  (2.8-26.1)  (7.9-22.3)
    (6 bases)
    GG(GG)1GG 11.2 +/− 9.1  15.1 +/− 10.0 13.8 +/− 6.5 
    SEQ ID NO:44-  (3.9-24.3)  (2.9-25.9)  (7.6-21.8)
    (6 bases)
    GG(GA)1GG 9.9 +/− 9.2 15.9 +/− 10.7 14.5 +/− 6.9 
    SEQ ID NO:45-  (2.6-23.1)  (2.9-26.9)  (7.9-23.0)
    (6 bases)
    GG(GC)1GG 4.7 +/− 3.4 15.8 +/− 10.4 14.1 +/− 6.6 
    SEQ ID NO:46- (1.7-9.3)  (3.0-26.2)  (8.3-21.7)
    (6 bases)
  • As shown in Table 41, 6 base GT, AGT, CGT, AG, CG and GG sequences increased chimpanzee PBMC cell production of the cytokines IL-10 and IL-12 and TNF-alpha.
  • EXAMPLE 37
  • Cytokine Synthesis by Rhesus Monkey Peripheral Blood Mononuclear Cells
  • PBMCs were isolated from 4 healthy rhesus monkeys as in Example 35. PBMCs were incubated with 6 base GT, AGT, CGT, AG, CG ands GG sequences and production of the cytokines IL-6, IL-12 and TNF-alpha was determined (Table 42).
    TABLE 42
    Cytokine production by rhesus monkeys PBMC
    IL-10 IL-12 TNF-alpha
    fold increase: fold increase: fold increase:
    mean +/− SD mean +/− SD mean +/− SD
    SEQUENCES (range) (range) (range)
    TG(TG)1TG 1.1 +/− 0.2 10.6 +/− 4.2  14.3 +/− 6.3 
    SEQ ID NO:9- (0.9-1.3)  (5.6-14.3)  (6.2-21.2)
    (6 bases)
    GT(GT)1GT 1.3 +/− 0.1 10.7 +/− 4.1  16.1 +/− 4.2 
    SEQ ID NO:10- (1.1-1.4)  (6.1-15.8) (11.0-21.6)
    (6 bases)
    TT(TG)1TT 1.0 +/− 0.1 6.7 +/− 3.6 4.4 +/− 3.8
    SEQ ID NO:23- (0.8-1.0)  (3.6-11.7) (1.3-9.5)
    (6 bases)
    GG(TG)1GG 1.0 +/− 0.1 11.2 +/− 4.8 14.5 +/− 5.4 
    SEQ ID NO:24- (1.0-1.1)  (5.6-16.8)  (7.7-20.0)
    (6 bases)
    GG(GT)1GG 1.0 +/− 0.1 10.6 +/− 4.7  12.5 +/− 5.2 
    SEQ ID NO:25- (1.0-1.1)  (5.5-16.5)  (6.2-18.6)
    (6 bases)
    TT(GT)1TT 1.0 +/− 0.1 4.9 +/− 2.9 2.1 +/− 1.0
    SEQ ID NO:26- (1.0-1.2) (2.6-8.8) (1.3-3.4)
    (6 bases)
    AA(GT)1AA 0.9 +/− 0.1 7.6 +/− 2.6 6.0 +/− 5.0
    SEQ ID NO:27- (0.9-1.0)  (5.9-11.5)  (2.4-13.4)
    (6 bases)
    CC(GT)1CC 1.1 +/− 0.2 10.1 +/− 3.7  14.2 +/− 3.7 
    SEQ ID NO:28- (0.9-1.2)  (6.2-14.2)  (9.6-18.6)
    (6 bases)
    TG(GT)1TG 1.1 +/− 0.2 11.1 +/− 4.2  16.5 +/− 2.7 
    SEQ ID NO:29- (0.9-1.2)  (6.5-15.7) (14.0-19.1)
    (6 bases)
    AT(GT)1AT 1.9 +/− 1.4 6.0 +/− 1.9 6.9 +/− 4.7
    SEQ ID NO:30- (1.0-4.0) (5.6-8.5)  (2.2-13.4)
    (6 bases)
    CT(GT)1CT 1.0 +/− 0.1 10.7 +/− 4.6  15.3 +/− 3.6 
    SEQ ID NO:36- (0.9-1.1)  (6.2-16.4) (11.2-19.8)
    (6 bases)
    TC(GT)1TC 1.1 +/− 0.2 10.0 +/− 3.8  14.7 +/− 3.1 
    SEQ ID NO:37- (0.9-1.3)  (6.3-14.1) (13.7-19.1)
    (6 bases)
    GG(TT)1GG 1.9 +/− 1.5 11.5 +/− 5.9  14.1 +/− 8.2 
    SEQ ID NO:41- (1.0-4.1)  (4.3-17.2)  (2.5-20.7)
    (6 bases)
    GG(AA)1GG 1.2 +/− 0.2 11.9 +/− 5.4  16.5 +/− 4.5 
    SEQ ID NO:42- (1.0-1.4)  (5.9-17.1) (11.4-21.2)
    (6 bases)
    GG(CC)1GG 1.0 +/− 0.2 11.7 +/− 4.8  16.9 +/− 3.5 
    SEQ ID NO:43- (0.8-1.2)  (6.2-16.4) (13.9-21.1)
    (6 bases)
    GG(GG)1GG 2.1 +/− 1.0 10.7 +/− 5.6  13.9 +/− 8.5 
    SEQ ID NO:44- (1.1-3.5)  (3.7-15.9)  (2.0-20.9)
    (6 bases)
    GG(GA)1GG 1.1 +/− 0.1 11.9 +/− 4.5  16.7 +/− 4.6 
    SEQ ID NO:45- (1.0-1.3)  (6.8-16.0) (11.6-21.5)
    (6 bases)
    GG(GC)1GG 1.2 +/− 0.2 11.0 +/− 4.4  16.8 +/− 4.1 
    SEQ ID NO:46- (1.0-1.4)  (6.3-16.1) (11.9-21.8)
    (6 bases)

    As shown in Table 42, 6 base GT, AGT, CGT, AG, CG and GG sequences increased rhesus monkey PBMC cell production of the cytokines IL-10, IL-12 and TNF-alpha.
  • EXAMPLE 38
  • Effect of 6 Base GT SEQ ID NO:25 of 6 Base GT SEQ ID NO:25+5-Fluorouracil and of 6 Base GT SEQ ID NO:25+Tamoxifen on MCF-7 Human Breast Tumors
  • MCF-7 human breast cancer cells are implanted subcutaneously as xenografts, in female nude BALB/c mice. The mice are divided into 6 groups of 10 mice. On day 0, group 1 mice receive saline, group 2 mice receive 6 base GT SEQ ID NO:25, group 3 mice receive receive 5-fluorouracil, group 4 mice receive tamoxifen, group 5 mice receive 6 base GT SEQ ID NO:25+5-fluorouracil and group 6 mice receive 6 base GT SEQ ID NO:25+tamoxifen. After 4 weeks of treatment, the mice are sacrificed and tumor mass is determined. Group 1 mice have the most tumor mass, groups 2, 3 and 4 mice have less tumor mass than group 1 mice and groups 5 and 6 mice have less tumor mass than groups 1, 2, 3 and 4 mice.
  • EXAMPLE 39
  • Effect of 3 and 6 Base GT Sequences and 45 Base BCG A-3 Sequence on LNCaP Human Prostate Cancer Tumors
  • LNCaP human prostate cancer cells are implanted subcutaneously, as xenografts, in male nude nu/nu mice. The mice are divided into 5 groups of 10 mice. On day 0, group 1 mice receive saline, group 2 mice receive 3 base SEQ ID NO:8, group 3 mice receive 6 base GT SEQ ID NO:25, group 4 mice receive 6 base AG SEQ ID NO:45 and group 5 mice receive 45 base BCG A-3 SEQ ID NO:69. After 4 weeks of treatment, the mice are sacrificed and timor mass is determined. Group 1 mice have the most tumor mass, group 5 mice have less tumor mass than group 1 mice and groups 2, 3 and 4 mice have less tumor mass than groups 1 and 5 mice.
  • EXAMPLE 41
  • Effect of 3, 6, 8 and 27 Base Sequences on EL-4 Murine T lymphomas
  • EL-4 murine T lymphoma cells are implanted into C57/BL6 mice. The mice are divided into 6 groups of 10 mice. On day 0, group 1 mice receive saline, group 2 mice receive 3 base GT SEQ ID NO:8, group 3 mice receive 6 base SEQ ID NO:25, group 4 mice receive 6 base AG SEQ ID NO:45, group 5 mice receive 18 base GT SEQ ID NO: 18 and group 6 mice receive 27 base GT SEQ ID NO:1. After 4 weeks of treatment, the mice are sacrificed and tumor mass is determined. Group 1 mice have the most tumor mass, groups 2, 3, 4, 5 and 6 mice have less tumor mass than group 1 mice.
  • EXAMPLE 42
  • Human colon cancer cell lines are maintained as adherent cell cultures. Cells in the exponential growth phase are treated with 2-20 base GT, GA, GC or GG sequences in the dose range 0 μg/ml to 100 μl/ml for 24-72 hours. Inhibition of cell proliferation is measured by MTT reduction, cell cycle arrest by flow cytometry and apoptosis by annexin-V binding or NuMA release. GT, GA, GC or GG sequences inhibit proliferation, induce cell cycle arrest and induce apoptosis in the colon cancer cell lines.
  • SCID mice bearing subcutaneous human colorectal cancer cell lines are treated with saline or with 2-20 base GT, GA, GC or GG sequences, having anti cancer activity against human colorectal cancer cell lines in vitro- Mice treated with 2-20 base GT, GA, GC or GG sequences, having anti-cancer activity against human colorectal cancer cell lines in vitro, show a significant reduction in tumor mass compared with mice treated with saline.
  • While the invention has been described in detail and with reference to specific embodiments thereof, it will be apparent to one skilled in the art that various changes and modifications can be made therein without departing from the spirit and scope thereof.

Claims (42)

1. A composition, comprising a 2 to 20 base 3′-OH, 5′-OH synthetic sequence selected from the group consisting of (GxTy)n, (TyGx)n, a(GxTy)n, a(TyGx)n, (GxTy)nb, (TyGx)nb, a(GxTy)nb, a(TyGx)nb, wherein x and y is an integer between 1 and 7, n is an integer between 1 and 12, a and b are one or more As, Cs, Gs or Ts and a pharmaceutically acceptable carrier.
2. The composition of claim 1, wherein the sequence is between 2 and 15 bases.
3. The composition of claim 2, wherein the sequence is between 2 and 10 bases.
4. The composition of claim 3, wherein the sequence is between 2 and 7 bases.
5. The composition of claim 1, further comprising a chemotherapeutic agent.
6. The composition of claim 5, wherein the chemotherapeutic agent is selected from the group consisting of antimetabolites, alkylating agents and hormone antagonists.
7. A composition, comprising a sequence selected from the group consisting SEQ ID NOs:7-18, 23-47, 50-66, and 81-83 and a pharmaceutically acceptable carrier.
8. The composition of claim 7, wherein the sequence is selected from the group consisting of SEQ ID NOs:7, 8, 9, 10, 22-65, 70-75, 79 and-80.
9. The composition of claim 7, further comprising a chemotherapeutic agent.
10. The composition of claim 9, wherein the chemotherapeutic agent is selected from the group consisting of an antimetabolite, an alkylating agent and an hormone antagonist.
11. A method, wherein a composition comprising a 2-20 base 3′-OH, 5′-OH synthetic sequence selected from the group consisting of (GxTy)n, (TyGx)n, a(GxTy)n, a(TyGx)n, (GxTy)nb, (TyGx)nb, a(GxTy)nb, a(TyGx)nb, wherein x and y is an integer between 1 and 7, n is an integer between 1 and 12, a and b are one or more As, Cs, Gs or Ts and a pharmaceutically acceptable carrier is administered to an animal having cancer in an amount effective to induce a response selected from the group consisting of induction of cell cycle arrest, inhibition of proliferation, activation of caspases and induction of apoptosis in cancer cells and production of cytokines by immune system cells.
12. The method of claim 11, wherein the sequence is between 2 and 15 bases.
13. The method of claim 12, wherein the sequence is between 2 and 10 bases.
14. The method of claim 13, wherein the sequence is between 2 and 7 bases.
15. The method of claim 11, wherein the response is induction of cell cycle arrest in the cancer cells.
16. The method of claim 11, wherein the response is inhibition of proliferation of the cancer cells.
17. The method of claim 11, wherein the response is activation of caspases in the cancer cells.
18. The method of claim 17, wherein the caspases are selected from the group consisting of caspase 3 and caspase 7.
19. The method of claim 11, wherein the response is induction of apoptosis in the cancer cells.
20. The method of claim 19, wherein the induction of apoptosis is independent of a cell property selected from the group consisting of Fas, p53/p21, p21/waf-1/CIP, p15ink4B, p16ink4, drug resistance, caspase 3, TGF-beta 1 receptor and hormone dependence.
21. The method of claim 11, wherein the response is production of cytokines by the immune system cells.
22. The method of claim 21, wherein the cytokines are selected from the group consisting of IL-1-beta, IL-6, IL-10, IL-12, and TNF-alpha.
23. The method of claim 11, wherein the cancer is selected from the group consisting of a primary carcinoma, a secondary carcinoma, a primary sarcoma and a secondary sarcoma.
24. The method of claim 23, wherein the cancer is selected from the group consisting of leukemia, lymphoma, breast, prostate, colorectal, ovarian and bone cancer and metastases therefrom.
25. The method of claim 11, further comprising a chemotherapeutic agent.
26. The method of claim 25, wherein the chemotherapeutic agent is selected from the group consisting of an antimetabolite, an alkylating agent and an hormone antagonists.
27. A method, wherein a composition comprising a 2-20 base 3′-OH, 5″-OH synthetic sequence selected from the group consisting SEQ ID NOs:7-18, 23-47, 50-66, and 81-83 and a pharmaceutically acceptable carrier is administered to an animal having cancer in an amount effective to induce a response selected from the group consisting of induction of cell cycle arrest, inhibition of proliferation, activation of caspases and induction of apoptosis in cancer cells and production of cytokines by immune system cells.
28. The method of claim 27, wherein the sequences are selected from the group consisting of SEQ ID NOs:7, 8, 9, 10, 22-65, 70-75, 79 and 80.
29. The method of claim 27, wherein the response is induction of cell cycle arrest in the cancer cells.
30. The method of claim 27, wherein the response is inhibition of proliferation of the cancer cells.
31. The method of claim 27, wherein the response is activation of caspases in the cancer cells.
32. The method of claim 31, wherein the caspases are selected from the group consisting of caspase 3 and caspase 7.
33. The method of claim 27, wherein the response is induction of apoptosis in the cancer cells.
34. The method of claim 33, wherein the induction of apoptosis is independent of a cell property selected from the group consisting of Fas, p53/p21, p21/waf-1/CIP, p155ink4B, p16ink4, drug resistance, caspase 3, TGF-beta 1 receptor and hormone dependence.
35. The method of claim 27, wherein the response is production of cytokines by the immune system cells.
36. The method of claim 35, wherein the cytokines are selected from the group consisting of IL-1-beta, IL-6, IL-10, IL-12, and TNF-alpha.
37. The method of claim 27, wherein the cancer is selected from the group consisting of a primary carcinoma, a secondary carcinoma, a primary sarcoma and a secondary sarcoma.
38. The method of claim 37, wherein the cancer is selected from the group consisting of leukemia, lymphoma, breast, prostate, colorectal, ovarian and bone cancer and metastases therefrom.
39. The method of claim 27, further comprising a chemotherapeutic agent.
40. The method of claim 39, wherein the chemotherapeutic agent is selected from the group consisting of an antimetabolite, an alkylating agent and an hormone antagonists.
41. A method, wherein a composition comprising a 2-20 base 3′-OH, 5′-OH synthetic sequence selected from the group consisting of (GxTy)n, (TyGx)n, a(GxTy)n, a(TyGx)n, (GxTy)nb, (Tyl G x)nb, a(GxTy)nb, a(TyGx)nb, wherein x and y is an integer between 1 and 7, n is an integer between 1 and 12, a and b are one or more As, Cs, Gs or Ts and a pharmaceutically acceptable carrier is administered to an animal having cancer in an amount effective to treat the cancer in the animal.
42. A method, wherein a composition comprising a sequence selected from the group consisting of SEQ ID NOs:7-18, 23-47, 50-66, and 81-83 and a pharmaceutically acceptable carrier is administered to an animal having cancer in an amount effective to treat the cancer in the animal.
US11/641,542 1999-12-13 2006-12-18 Therapeutically useful synthetic oligonucleotides Abandoned US20070213291A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/641,542 US20070213291A1 (en) 1999-12-13 2006-12-18 Therapeutically useful synthetic oligonucleotides

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US17032599P 1999-12-13 1999-12-13
US22892500P 2000-08-29 2000-08-29
US09/735,363 US7157436B2 (en) 1999-12-13 2000-12-12 Therapeutically useful synthetic oligonucleotides
US11/641,542 US20070213291A1 (en) 1999-12-13 2006-12-18 Therapeutically useful synthetic oligonucleotides

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/735,363 Continuation US7157436B2 (en) 1999-12-13 2000-12-12 Therapeutically useful synthetic oligonucleotides

Publications (1)

Publication Number Publication Date
US20070213291A1 true US20070213291A1 (en) 2007-09-13

Family

ID=26865979

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/735,363 Expired - Fee Related US7157436B2 (en) 1999-12-13 2000-12-12 Therapeutically useful synthetic oligonucleotides
US11/641,542 Abandoned US20070213291A1 (en) 1999-12-13 2006-12-18 Therapeutically useful synthetic oligonucleotides

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/735,363 Expired - Fee Related US7157436B2 (en) 1999-12-13 2000-12-12 Therapeutically useful synthetic oligonucleotides

Country Status (18)

Country Link
US (2) US7157436B2 (en)
EP (2) EP1238070B1 (en)
JP (1) JP4772245B2 (en)
KR (1) KR100829646B1 (en)
CN (2) CN100445380C (en)
AT (2) ATE455851T1 (en)
AU (1) AU785212B2 (en)
CA (1) CA2393808A1 (en)
CZ (1) CZ20022372A3 (en)
DE (2) DE60036019T2 (en)
DK (2) DK1238070T3 (en)
ES (2) ES2340807T3 (en)
HU (1) HUP0300627A3 (en)
IL (2) IL150196A0 (en)
MX (1) MXPA02005842A (en)
NO (1) NO330508B1 (en)
PT (1) PT1238070E (en)
WO (1) WO2001044465A2 (en)

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030032610A1 (en) 1996-06-03 2003-02-13 Gilchrest Barbara A. Method to inhibit cell growth using oligonucleotides
US8114850B2 (en) 1999-04-08 2012-02-14 Advanced Cancer Therapeutics, Llc Antiproliferative activity of G-rich oligonucleotides and method of using same to bind to nucleolin
DE60036700T2 (en) 1999-04-08 2008-07-24 Antisoma Research Ltd. ANTIPROLIFERATIVE ACTIVITY OF G-rich OLIGONUCLEOTIDES AND METHOD TO USE THEM TO BIND NUCLEOTINE
US7960540B2 (en) 1999-04-08 2011-06-14 Advanced Cancer Therapeutics, Llc Antiproliferative activity of G-rich oligonucleotides and method of using same to bind to nucleolin
US6949520B1 (en) * 1999-09-27 2005-09-27 Coley Pharmaceutical Group, Inc. Methods related to immunostimulatory nucleic acid-induced interferon
AU785212B2 (en) * 1999-12-13 2006-11-09 Bioniche Life Sciences Inc. Therapeutically useful synthetic oligonucleotides
EP1313853B1 (en) 2000-08-29 2012-03-21 Bioniche Life Sciences Inc. Modulation of fas and fasl expression by a synthetic phosphodiester oligonucleotide and an anti-fas antibody
ATE398175T1 (en) * 2000-12-08 2008-07-15 Coley Pharmaceuticals Gmbh CPG-TYPE NUCLEIC ACIDS AND METHODS OF USE THEREOF
US7087586B2 (en) * 2001-04-24 2006-08-08 Bioniche Life Sciences, Inc. Oligonucleotide compositions and their use to induce differentiation of cells
DK1417307T3 (en) * 2001-08-17 2009-07-13 Bioniche Life Sciences Inc Oligonucleotide Compositions and Their Use to Induce Apoptosis
MXPA04002530A (en) 2001-10-03 2004-05-31 Bioniche Life Sciences Inc Therapeutically useful triethyleneglycol cholesteryl oligonucleotides.
JP2006515266A (en) * 2002-04-22 2006-05-25 バイオニケ ライフ サイエンシーズ インコーポレイテッド Oligonucleotide compositions and their use for modulating immune responses
CA2521050A1 (en) * 2003-04-02 2004-10-14 Coley Pharmaceutical Group, Ltd. Immunostimulatory nucleic acid oil-in-water formulations and related methods of use
WO2004103301A2 (en) * 2003-05-16 2004-12-02 Hybridon, Inc. Synergistic treatment of cancer using immunomers in conjunction with chemotherapeutic agents
CA2598992A1 (en) * 2005-02-24 2006-08-31 Coley Pharmaceutical Group, Inc. Immunostimulatory oligonucleotides
WO2006099021A2 (en) * 2005-03-09 2006-09-21 Baylor College Of Medicine Direct reversal of the suppressive function of cd4+ regulatory t cells via toll-like receptor 8 signaling
WO2007028985A2 (en) * 2005-09-07 2007-03-15 The Secretary Of State For Defence Adjuvanted vaccine
KR101805199B1 (en) * 2009-02-12 2017-12-05 큐알엔에이, 인크. Treatment of glial cell derived neurotrophic factor (gdnf) related diseases by inhibition of natural antisense transcript to gdnf
GB0906234D0 (en) 2009-04-14 2009-05-20 Secr Defence Vaccine
KR100998365B1 (en) 2009-06-29 2010-12-06 압타바이오 주식회사 Novel guanosine rich modified oligonucleotides and antiproliferative activity thereof
US9040493B2 (en) * 2009-07-15 2015-05-26 Santaris Pharma A/S RNA antagonists targeting GLI2 for the treatment of leukemia
WO2013129603A1 (en) * 2012-02-28 2013-09-06 国立大学法人東京農工大学 Method for designating disease relating to amount of tdp-43 existing in cells
EP4000597A1 (en) * 2020-11-17 2022-05-25 The Boots Company plc Tetrapeptide and compositions comprising tetrapeptides
WO2023122762A1 (en) * 2021-12-22 2023-06-29 Camp4 Therapeutics Corporation Modulation of gene transcription using antisense oligonucleotides targeting regulatory rnas

Citations (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3172815A (en) * 1962-04-24 1965-03-09 Warner Lambert Pharmaceutical Method for preparing reticulo-endo-thelial system stimulant and stimulant thereof
US3976544A (en) * 1973-06-19 1976-08-24 The Agence Nationale De Valorisation De Le Recherche Water-soluble immunological adjuvants, in particular for vaccines, obtained from mycobacteria and related microorganisms and process for their extraction
US4010257A (en) * 1973-02-23 1977-03-01 Burroughs Wellcome Co. Biological extracts
US4036953A (en) * 1971-11-19 1977-07-19 Agence Nationale De Valorisation De La Recherche (Anvar) Process for preparing vaccine adjuvant
US4152423A (en) * 1971-11-19 1979-05-01 Agence Nationale De Valorisation De La Recherche (Anvar) Immunological agents
US4182751A (en) * 1976-06-04 1980-01-08 Institut Merieux New immunostimulant medicament and process of preparing same
US4520019A (en) * 1982-04-29 1985-05-28 Ribi Immunochem Research, Inc. Stable composition and preparation thereof
US4579941A (en) * 1982-08-13 1986-04-01 Mitsuitoatsu Chemicals, Inc. Thermally-denatured deoxyribonucleic acid MD-011 and antitumor agent
US4647456A (en) * 1980-03-25 1987-03-03 Dr. Madaus & Co. Methods of increasing tolerance to radiotherapy and chemotherapy using propioni bacteria
US4663306A (en) * 1983-09-23 1987-05-05 Ribi Immunochem Research, Inc. Pyridine-soluble extract-refined detoxified endotoxin composition and use
US4724144A (en) * 1984-02-17 1988-02-09 University College London Immuno-therapeutic composition of killed cells from mycobacterium vaccae
US4726947A (en) * 1978-03-10 1988-02-23 Matsui Toatsu Chemicals, Inc. Bacterial cell extract, process for preparing same, antitumor preparation containing same, and adjuvant preparation containing same
US4744984A (en) * 1985-10-08 1988-05-17 Vetrepharm Research, Inc. Antiviral immunotherapeutic agent and preparation thereof
US4791101A (en) * 1984-06-23 1988-12-13 Boehringer Ingelheim Synergistic mixtures of interferons and tumor necrosis factor
US4877611A (en) * 1986-04-15 1989-10-31 Ribi Immunochem Research Inc. Vaccine containing tumor antigens and adjuvants
US4958013A (en) * 1989-06-06 1990-09-18 Northwestern University Cholesteryl modified oligonucleotides
US4983518A (en) * 1987-07-11 1991-01-08 Boehringer Mannheim Gmbh Eukaryotic expression vectors with multimeric enhancer segments, a process for the preparation thereof and the use thereof
US5474796A (en) * 1991-09-04 1995-12-12 Protogene Laboratories, Inc. Method and apparatus for conducting an array of chemical reactions on a support surface
US5582981A (en) * 1991-08-14 1996-12-10 Gilead Sciences, Inc. Method for identifying an oligonucleotide aptamer specific for a target
US5643890A (en) * 1995-01-31 1997-07-01 The Board Of Regents Of The University Of Nebraska Synthetic oligonucleotides which mimic telomeric sequences for use in treatment of cancer and other diseases
US5759554A (en) * 1993-01-29 1998-06-02 Vetrepharm, Inc. Immunostimulatory bacterial cell wall traction
US5827834A (en) * 1989-09-21 1998-10-27 Hyal Pharmaceutical Corporation Method of using hyaluronic acid or its pharmaceutically acceptable salts for the treatment of disease
US5861245A (en) * 1990-10-15 1999-01-19 Stratagene & California Institute Of Biological Research Arbitrarily primed polymerase chain reaction method for fingerprinting genomes
US5888986A (en) * 1995-02-14 1999-03-30 Bioniche Inc. Method for treating the urinary bladder and associated structures using hyaluronic acid
US6015710A (en) * 1996-04-09 2000-01-18 The University Of Texas System Modulation of mammalian telomerase by peptide nucleic acids
US6150339A (en) * 1993-04-23 2000-11-21 Aronex Pharmaceuticals, Inc. Anti-viral guanosine-rich oligonucleotides
US6211431B1 (en) * 1994-08-30 2001-04-03 Commonwealth Scientific And Industrial Research Organization Plant transcription regulators from circovirus
US6255473B1 (en) * 1997-08-29 2001-07-03 Duke University Presenilin-1 gene promoter
US6316190B1 (en) * 1996-05-20 2001-11-13 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Oligonucleotides which specifically bind retroviral nucleocapsid proteins
US20010041681A1 (en) * 1999-12-13 2001-11-15 Phillips Nigel C. Therapeutically useful synthetic oligonucleotides
US6326357B1 (en) * 1997-08-05 2001-12-04 Bioniche Life Sciences Inc. Bacterial cell complex composition and method of use
US20020091095A1 (en) * 1999-12-13 2002-07-11 Phillips Nigel C. Modulation of Fas and FasL expression
US6475795B1 (en) * 1991-07-03 2002-11-05 Meditech Research, Ltd. Use of hyaluronan in gene therapy
US20030125290A1 (en) * 2001-10-03 2003-07-03 Phillips Nigel C. Therapeutically useful triethyleneglycol cholesteryl oligonucleotides
US7125858B2 (en) * 1999-12-28 2006-10-24 Bioniche Life Sciences Inc. Hyaluronic acid in the treatment of cancer
US20070066554A1 (en) * 1999-09-25 2007-03-22 Coley Pharmaceutical Gmbh Immunostimulatory nucleic acids
US7371734B2 (en) * 2002-04-22 2008-05-13 Bioniche Life Sciences Inc. Oligonucleotide compositions and their use for the modulation of immune responses

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992014842A1 (en) * 1991-02-21 1992-09-03 Gilead Sciences, Inc. Aptamers specific for thrombin and methods of use
US5523389A (en) 1992-09-29 1996-06-04 Isis Pharmaceuticals, Inc. Inhibitors of human immunodeficiency virus
CN1154702A (en) * 1994-05-25 1997-07-16 海布里登公司 Oligonucleotides with anti-cytomegalovirus activity
ZA964446B (en) * 1995-06-06 1996-12-06 Hoffmann La Roche Oligonucleotides specific for hepatitis c virus
US5990299A (en) * 1995-08-14 1999-11-23 Icn Pharmaceuticals, Inc. Control of CD44 gene expression for therapeutic use
SK66098A3 (en) * 1995-11-21 1998-11-04 Icn Pharmaceuticals Inhibition of tumor growth by antisense oligonucleotides for il-8 and il-8 receptor
IT1277025B1 (en) * 1995-12-04 1997-11-04 Cooperativa Centro Ricerche Po CLASS OF PHOSPHODIESTERIC OLIGONUCLEOTIDES WITH CYTOTOXIC ACTIVITY PHARMACEUTICAL COMPOSITIONS THAT CONTAIN THEM AND THEIR USE

Patent Citations (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3172815A (en) * 1962-04-24 1965-03-09 Warner Lambert Pharmaceutical Method for preparing reticulo-endo-thelial system stimulant and stimulant thereof
US4036953A (en) * 1971-11-19 1977-07-19 Agence Nationale De Valorisation De La Recherche (Anvar) Process for preparing vaccine adjuvant
US4152423A (en) * 1971-11-19 1979-05-01 Agence Nationale De Valorisation De La Recherche (Anvar) Immunological agents
US4010257A (en) * 1973-02-23 1977-03-01 Burroughs Wellcome Co. Biological extracts
US3976544A (en) * 1973-06-19 1976-08-24 The Agence Nationale De Valorisation De Le Recherche Water-soluble immunological adjuvants, in particular for vaccines, obtained from mycobacteria and related microorganisms and process for their extraction
US4182751A (en) * 1976-06-04 1980-01-08 Institut Merieux New immunostimulant medicament and process of preparing same
US4337243A (en) * 1976-06-04 1982-06-29 Institut Merieux, S.A. Immunostimulant medicament and process of preparing same
US4726947A (en) * 1978-03-10 1988-02-23 Matsui Toatsu Chemicals, Inc. Bacterial cell extract, process for preparing same, antitumor preparation containing same, and adjuvant preparation containing same
US4647456A (en) * 1980-03-25 1987-03-03 Dr. Madaus & Co. Methods of increasing tolerance to radiotherapy and chemotherapy using propioni bacteria
US4520019A (en) * 1982-04-29 1985-05-28 Ribi Immunochem Research, Inc. Stable composition and preparation thereof
US4579941A (en) * 1982-08-13 1986-04-01 Mitsuitoatsu Chemicals, Inc. Thermally-denatured deoxyribonucleic acid MD-011 and antitumor agent
US4663306A (en) * 1983-09-23 1987-05-05 Ribi Immunochem Research, Inc. Pyridine-soluble extract-refined detoxified endotoxin composition and use
US4724144A (en) * 1984-02-17 1988-02-09 University College London Immuno-therapeutic composition of killed cells from mycobacterium vaccae
US4791101A (en) * 1984-06-23 1988-12-13 Boehringer Ingelheim Synergistic mixtures of interferons and tumor necrosis factor
US4744984A (en) * 1985-10-08 1988-05-17 Vetrepharm Research, Inc. Antiviral immunotherapeutic agent and preparation thereof
US4877611A (en) * 1986-04-15 1989-10-31 Ribi Immunochem Research Inc. Vaccine containing tumor antigens and adjuvants
US4983518A (en) * 1987-07-11 1991-01-08 Boehringer Mannheim Gmbh Eukaryotic expression vectors with multimeric enhancer segments, a process for the preparation thereof and the use thereof
US4958013A (en) * 1989-06-06 1990-09-18 Northwestern University Cholesteryl modified oligonucleotides
US5827834A (en) * 1989-09-21 1998-10-27 Hyal Pharmaceutical Corporation Method of using hyaluronic acid or its pharmaceutically acceptable salts for the treatment of disease
US5861245A (en) * 1990-10-15 1999-01-19 Stratagene & California Institute Of Biological Research Arbitrarily primed polymerase chain reaction method for fingerprinting genomes
US6475795B1 (en) * 1991-07-03 2002-11-05 Meditech Research, Ltd. Use of hyaluronan in gene therapy
US5582981A (en) * 1991-08-14 1996-12-10 Gilead Sciences, Inc. Method for identifying an oligonucleotide aptamer specific for a target
US5474796A (en) * 1991-09-04 1995-12-12 Protogene Laboratories, Inc. Method and apparatus for conducting an array of chemical reactions on a support surface
US5759554A (en) * 1993-01-29 1998-06-02 Vetrepharm, Inc. Immunostimulatory bacterial cell wall traction
US6150339A (en) * 1993-04-23 2000-11-21 Aronex Pharmaceuticals, Inc. Anti-viral guanosine-rich oligonucleotides
US6211431B1 (en) * 1994-08-30 2001-04-03 Commonwealth Scientific And Industrial Research Organization Plant transcription regulators from circovirus
US5643890A (en) * 1995-01-31 1997-07-01 The Board Of Regents Of The University Of Nebraska Synthetic oligonucleotides which mimic telomeric sequences for use in treatment of cancer and other diseases
US5888986A (en) * 1995-02-14 1999-03-30 Bioniche Inc. Method for treating the urinary bladder and associated structures using hyaluronic acid
US6015710A (en) * 1996-04-09 2000-01-18 The University Of Texas System Modulation of mammalian telomerase by peptide nucleic acids
US6316190B1 (en) * 1996-05-20 2001-11-13 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Oligonucleotides which specifically bind retroviral nucleocapsid proteins
US6326357B1 (en) * 1997-08-05 2001-12-04 Bioniche Life Sciences Inc. Bacterial cell complex composition and method of use
US6329347B1 (en) * 1997-08-05 2001-12-11 Bioniche Life Sciences Inc. Composition and method for the treatment of bladder cancer
US6255473B1 (en) * 1997-08-29 2001-07-03 Duke University Presenilin-1 gene promoter
US20070066554A1 (en) * 1999-09-25 2007-03-22 Coley Pharmaceutical Gmbh Immunostimulatory nucleic acids
US20020091095A1 (en) * 1999-12-13 2002-07-11 Phillips Nigel C. Modulation of Fas and FasL expression
US20030119776A1 (en) * 1999-12-13 2003-06-26 Phillips Nigel C. Modulation of Fas and FasL expression
US7157436B2 (en) * 1999-12-13 2007-01-02 Bioniche Life Sciences, Inc. Therapeutically useful synthetic oligonucleotides
US20010041681A1 (en) * 1999-12-13 2001-11-15 Phillips Nigel C. Therapeutically useful synthetic oligonucleotides
US7125858B2 (en) * 1999-12-28 2006-10-24 Bioniche Life Sciences Inc. Hyaluronic acid in the treatment of cancer
US20030125290A1 (en) * 2001-10-03 2003-07-03 Phillips Nigel C. Therapeutically useful triethyleneglycol cholesteryl oligonucleotides
US7371734B2 (en) * 2002-04-22 2008-05-13 Bioniche Life Sciences Inc. Oligonucleotide compositions and their use for the modulation of immune responses

Also Published As

Publication number Publication date
DE60043759D1 (en) 2010-03-11
MXPA02005842A (en) 2003-10-14
EP1238070A2 (en) 2002-09-11
WO2001044465A2 (en) 2001-06-21
KR100829646B1 (en) 2008-05-16
AU785212B2 (en) 2006-11-09
KR20020079752A (en) 2002-10-19
EP1867718A3 (en) 2008-04-02
CN1433469A (en) 2003-07-30
DK1867718T3 (en) 2010-04-12
CN101491536B (en) 2012-07-04
ES2340807T3 (en) 2010-06-09
DE60036019D1 (en) 2007-09-27
ATE455851T1 (en) 2010-02-15
HUP0300627A3 (en) 2005-11-28
CN101491536A (en) 2009-07-29
US20010041681A1 (en) 2001-11-15
JP2003517041A (en) 2003-05-20
ATE370231T1 (en) 2007-09-15
CA2393808A1 (en) 2001-06-21
NO20022820D0 (en) 2002-06-13
EP1867718B1 (en) 2010-01-20
PT1238070E (en) 2007-11-06
AU1847901A (en) 2001-06-25
IL150196A (en) 2009-06-15
NO20022820L (en) 2002-08-08
NO330508B1 (en) 2011-05-09
EP1238070B1 (en) 2007-08-15
CN100445380C (en) 2008-12-24
DE60036019T2 (en) 2008-05-29
JP4772245B2 (en) 2011-09-14
DK1238070T3 (en) 2007-12-03
ES2288883T3 (en) 2008-02-01
EP1867718A2 (en) 2007-12-19
US7157436B2 (en) 2007-01-02
HUP0300627A2 (en) 2004-06-28
CZ20022372A3 (en) 2002-11-13
WO2001044465A3 (en) 2001-11-29
IL150196A0 (en) 2002-12-01

Similar Documents

Publication Publication Date Title
US20070213291A1 (en) Therapeutically useful synthetic oligonucleotides
EP1322655B1 (en) Oligodeoxynucleotide and its use to induce an immune response
ES2543710T3 (en) Immunostimulatory oligonucleotides containing G and U
Wang et al. Chemotherapy and chemosensitization of non–small cell lung cancer with a novel immunomodulatory oligonucleotide targeting Toll-like receptor 9
JP2009511034A (en) Methods and compositions for treating immune disorders
Gray et al. CpG-B ODNs potently induce low levels of IFN-αβ and induce IFN-αβ-dependent MHC-I cross-presentation in DCs as effectively as CpG-A and CpG-C ODNs
US7662792B2 (en) Modulation of Fas and FasL expression
KR101017483B1 (en) Oligonucleotide compositions and their use to induce apoptosis
AU2007200536A1 (en) Therapeutically useful synthetic oligonucleotides
EP1313853B1 (en) Modulation of fas and fasl expression by a synthetic phosphodiester oligonucleotide and an anti-fas antibody
ES2383671T3 (en) Modulation of Fas and FasL expression by a synthetic oligonucleotide phosphodiester and an anti-Fas antibody
AU2001268863B2 (en) Modulation of FAS and FASL expression
Tarhini et al. Early development of the toll-like receptor 9 agonist, PF-3512676, for the treatment of patients with advanced cancers
AU2001268863A1 (en) Modulation of FAS and FASL expression
AU2001268863A2 (en) Modulation of FAS and FASL expression
Gray Regulation of interferon alpha beta induction and dendritic cell function by CpG oligodeoxynucleotides
EP1950296A2 (en) Oligodeoxynucleotide and its use to induce an immune response

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION